EP2794854B1 - Nachweis von zellen aus menschlichem nabelschnurgewebe - Google Patents

Nachweis von zellen aus menschlichem nabelschnurgewebe Download PDF

Info

Publication number
EP2794854B1
EP2794854B1 EP12815943.1A EP12815943A EP2794854B1 EP 2794854 B1 EP2794854 B1 EP 2794854B1 EP 12815943 A EP12815943 A EP 12815943A EP 2794854 B1 EP2794854 B1 EP 2794854B1
Authority
EP
European Patent Office
Prior art keywords
cells
cell
umbilical cord
human
derived
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP12815943.1A
Other languages
English (en)
French (fr)
Other versions
EP2794854A1 (de
Inventor
Haimanti Dorai
Reyna L. FAVIS
Xiang Yao
Yu Sun
Anthony Kihm
Stefanie RASSNICK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DePuy Synthes Products Inc
Original Assignee
DePuy Synthes Products Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by DePuy Synthes Products Inc filed Critical DePuy Synthes Products Inc
Priority to PL12815943T priority Critical patent/PL2794854T3/pl
Publication of EP2794854A1 publication Critical patent/EP2794854A1/de
Application granted granted Critical
Publication of EP2794854B1 publication Critical patent/EP2794854B1/de
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6888Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for detection or identification of organisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70589CD45

Definitions

  • the invention relates to methods of detecting allogeneic therapeutic cells, such as e.g. human umbilical cord tissue-derived cells, in a sample from a patient that treated with the therapeutic cells.
  • allogeneic therapeutic cells such as e.g. human umbilical cord tissue-derived cells
  • Allogeneic cell therapies are a promising new technology for the treatment of a number of unmet medical needs.
  • cell therapies are unique products and pose some unique challenges in the development process.
  • One specific example of this technology is the development of human umbilical cord tissue-derived cells ("hUTC") for a number of clinical indications.
  • hUTC human umbilical cord tissue-derived cells
  • measuring the presence and/or the number of cells detected in the subject's blood is desirable information relevant to the pharmacokinetics of hUTC as a cell therapy product.
  • Clinical studies during drug development include pharmacokinetic studies to examine parameters of absorption, distribution, metabolism, and excretion of the drug in vivo.
  • An important element of the pharmacokinetic studies is to determine the level of exposure of a drug in subjects. Typically, this is done through analysis of drug levels from blood samples; exposure levels are evaluated relative to efficacy and safety outcomes.
  • studying the bio-distribution or pharmacokinetics of hUTC in clinical trials presents a challenge because there is no established method to distinguish hUTC (or other cell products) from the subjects' own cells. Therefore, it is difficult to determine the bioavailability of hUTC.
  • Real time PCR is used to detect the Y chromosome in a sample of the subject's blood and the results provide a relative quantification or binary signal to indicate the presence or absence of allogeneic therapeutic cells (e.g. hUTC) in the blood sample.
  • allogeneic therapeutic cells e.g. hUTC
  • This approach necessitates excluding female cell (e.g. hUTC) donors and male subjects from analyses on pharmacokinetics of hUTC in clinical studies. Therefore, there remains a need for a method for detecting allogeneic cells, such as e.g. UTC, in patients after administration of the cells.
  • the invention provides a method of detecting circulating allogeneic therapeutic cells in a blood sample comprising:
  • the invention further provides a method of detecting circulating human umbilical cord tissue-derived cells in a blood sample comprising the steps of:
  • the invention provides for detecting the presence of human umbilical cord tissue-derived cells in a sample of human peripheral blood mononuclear cells.
  • the invention allows for the detection of such therapeutic cells without being constrained by the therapeutic cells karyotype (XY vs. XX) and recipient (patient) gender.
  • the one or more positive markers for patient peripheral blood mononuclear cells comprises CD45.
  • the methods may be suitable to detect any allogeneic therapeutic cells of interest.
  • the allogeneic therapeutic cells may be selected from the group consisting of human umbilical cord tissue-derived cells, human umbilical cord blood-derived cells, placental-derived cells, mesenchymal stem cell derived cells, liver cells, pancreatic islet cells, cardiomyocytes, and insoluble collagenous bone matrix cells.
  • the methods may also be used to detect two or more different types of allogeneic therapeutic cells in a blood sample.
  • the methods may include an enrichment step between steps (a) and (b).
  • the enrichment step may include magnetic capture technology.
  • the step of distinguishing includes differentiating between allogeneic therapeutic cells administered to the patient and peripheral blood mononuclear cells from the patient.
  • the patient may be a human, non-human primate, mouse, rat, hamster, guinea pig, dog, or pig.
  • kits which may be used use in the method of detecting allogeneic therapeutic cells in a blood sample.
  • the kits may include a marker profile having one or more markers positive allogeneic therapeutic cells and one or more markers positive for patient peripheral blood mononuclear cells.
  • the patient may be a human, non-human primate, mouse, rat, hamster, guinea pig, dog, or pig.
  • the positive marker for patient peripheral blood mononuclear cells is CD45 and the positive marker for human umbilical cord tissue-derived cells is CD10.
  • markers for patient peripheral blood mononuclear cells and for human umbilical cord tissue-derived cells include one or more of CD10, CD13, NRP1, CD45, LAMP1, DKK3, NRP1, or LAMB1.
  • the method may employ a variety of assay techniques including flow cytometry, ELISA, immunohistochemistry, nucleic acid detection, PCR, and combinations thereof
  • the one or more unique markers positive for human umbilical cord tissue-derived cells may be one or more of CD10, CD13, NRP1, LAMP1, DKK3, NRP1, LAMB1 and combinations thereof.
  • the method may further comprise performing an enrichment step between steps (a) and (b).
  • the enrichment step may be magnetic capture technology.
  • Another embodiment of the invention is a method of detecting human umbilical cord tissue-derived cells in blood including the steps of: assaying human umbilical cord tissue-derived cells and human peripheral blood mononuclear cells to identify one or more markers positive for human umbilical cord tissue-derived cells and one or more markers positive for human peripheral blood mononuclear cells; providing a blood sample containing human umbilical cord tissue-derived cells; isolating the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction from the blood sample; analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD10 or CD13 as a positive marker for human umbilical cord tissue-derived cells and detecting the presence of the human peripheral blood mononuclear cells and human umbilical cord tissue-derived cells based on the detection of CD45 as a marker positive for human peripheral blood mononuclear cells and CD10 or CD13 as a marker
  • the step of analyzing may include analysis of the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD13 as a positive marker for human umbilical cord tissue-derived cells.
  • the step of analysis may include analysis of the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD10 as a positive marker for human umbilical cord tissue-derived cells.
  • the method may also further include performing an enrichment step prior to analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction such as e.g. magnetic capture technology.
  • Another embodiment of the invention is a method of detecting human umbilical cord tissue-derived cells in blood including the steps of: providing a blood sample containing human umbilical cord tissue-derived cells; isolating the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction from the blood sample; removing any plasma; analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD13 as a positive marker for human umbilical cord tissue-derived cells and detecting the presence of the human peripheral blood mononuclear cells and human umbilical cord tissue-derived cells based on the detection of CD45 as a marker positive for human peripheral blood mononuclear cells and CD13 as a marker positive for human umbilical cord tissue-derived cells.
  • the method may also further include performing an enrichment step prior to analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction such as e.g. magnetic capture technology.
  • the method may also further include detecting for CD10 as a marker positive for human umbilical cord tissue-derived cells.
  • Human umbilical cord tissue-derived cells are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, and have the potential to differentiate.
  • the human umbilical cord tissue-derived cells are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, and have the following characteristics: (1) express CD10, CD13, CD44, CD90, and HLA-ABC; (2) do not express CD31, CD34, CD45, HLA-DR and CD117, and (3) do not express hTERT or telomerase.
  • the human umbilical cord tissue-derived cells may be isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, and have the following characteristics: (1) express CD10, CD13, CD44, CD90, and HLA-ABC; (2) do not express CD31, CD34, CD45, HLA-DR and CD117; (3) do not express hTERT or telomerase; (4) express oxidized low density lipoprotein receptor 1, reticulon, chemokine receptor ligand 3, and/or granulocyte chemotactic protein; and (4) express, relative to a human fibroblast, mesenchymal stem cell, or iliac crest bone marrow cell, increased levels of interleukin 8 or reticulon 1.
  • Another embodiment of the invention is method of detecting human umbilical cord tissue-derived cells in blood including the steps of: (a) providing a blood sample from a patient that has been treated with human umbilical cord tissue-derived cells; (b) analyzing the sample using an assay method to detect one or more markers positive for human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells; and (c) distinguishing between the human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells.
  • the positive marker for human peripheral blood mononuclear cells is CD45 and the positive marker for human umbilical cord tissue-derived cells is CD10.
  • the step of analyzing may utilize flow cytometry, ELISA, immunohistochemistry, nucleic acid detection, and/or PCR.
  • the method may further include the performing an enrichment step between steps (a) and (b).
  • the enrichment step may be magnetic capture technology.
  • Yet another embodiment of the invention is a method of detecting human umbilical cord tissue-derived cells in blood including the steps of: (a) providing a blood sample containing human umbilical cord tissue-derived cells; (b) isolating the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction from the blood sample; and (c) analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD10 as a positive marker for human umbilical cord tissue-derived cells.
  • An alternate embodiment of the invention is a method of detecting human umbilical cord tissue-derived cells in blood comprising the steps of: providing a blood sample containing human umbilical cord tissue-derived cells; isolating the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction from the blood sample; removing any plasma; and analyzing the human umbilical cord tissue-derived cell/ peripheral blood mononuclear cell fraction by flow cytometry for CD45 as a positive marker for peripheral blood mononuclear cell and CD13 as a positive marker for human umbilical cord tissue-derived cells.
  • This application is directed to methods of detecting allogeneic therapeutic cells such as progenitor cells circulating in the blood of a patient (e.g. human).
  • Progenitor cells or other engineered cells which are components of a cell therapy product, are being developed for a number of clinical indications. These cells, such as e.g. hUTC, are administered to the patient via e.g. intravenous administration. The disposition of these cells in circulating blood needs to be determined in order to assess the pharmacokinetics of the cells as a cell therapy product and also to more accurately determine success of the therapy.
  • human blood is composed of multiple types of blood cells, one or more type of which express some proteins that may also be expressed in the progenitor or engineered cells.
  • sample refers to any substance, which may contain the analyte of interest.
  • a sample can be biological fluid, such as whole blood or whole blood components including red blood cells, white blood cells, platelets, serum and plasma, ascetic fluid, urine, cerebrospinal fluid, and other constituents of the body.
  • the cells which may be identified in a blood sample containing peripheral mononuclear cells using the methods of the disclosure are generally referred to as postpartum cells or postpartum-derived cells (PPDCs).
  • the cells are more specifically "umbilicus-derived cells” or “umbilical cord-derived cells” (UDC), or “umbilical cord tissue-derived cells” (UTC) or “human umbilical cord tissue-derived cells” (hUTC).
  • the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense.
  • the term "derived” is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a growth medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below.
  • Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • HSC hematopoietic stem cells
  • HSC hematopoietic stem cells
  • Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells.
  • Cells that are unipotent are able to give rise to a single cell lineage (e.g. , spermatogenic stem cells).
  • Stem cells are also categorized based on the source from which they are obtained.
  • An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types.
  • An embryonic stem cell is a pluripotent cell that can be derived from the inner cell mass of a blastocyst-stage embryo.
  • a fetal stem cell is one that originates from fetal tissues or membranes.
  • a postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the umbilical cord.
  • Postpartum stem cells may be blood-derived (e.g. , as are those obtained from umbilical cord blood) or non-blood-derived (e.g. , as obtained from the non-blood tissues of the umbilical cord).
  • Cell culture refers generally to cells taken from a living organism and grown under controlled conditions ("in culture” or “cultured”).
  • a “primary cell culture” is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. Cells are “expanded” in culture when they are placed in a growth medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells. When cells are expanded in culture, the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as "doubling time.”
  • cell line generally refers to a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been "passaged.” A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture.
  • the primary culture i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (PI or passage 1).
  • the cells After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore, the number of population doublings of a culture is greater than the passage number.
  • the expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including, but not limited to, the seeding density, substrate, medium, growth conditions, and time between passaging.
  • “Differentiation” is the process by which an unspecialized ("uncommitted") or less specialized cell acquires the features of a specialized cell, such as e.g. a nerve cell or a muscle cell.
  • a “differentiated” cell is one that has taken on a more specialized ("committed") position within the lineage of a cell.
  • the term “committed”, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type.
  • Dedifferentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell.
  • the "lineage" of a cell defines the heredity of the cell, i.e., which cells it came from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors.
  • stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells.
  • this broad definition of progenitor cell may be used.
  • a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types.
  • progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a “non-renewing progenitor cell” or as an “intermediate progenitor or precursor cell.”
  • a “trophic factor” is defined as a substance that promotes survival, growth, proliferation, and/or maturation of a cell, or stimulates increased activity of a cell.
  • standard growth conditions refers to culturing of cells at 37°C, in a standard atmosphere comprising 5% CO 2 and relative humidity maintained at about 100%. While the foregoing conditions are useful for culturing, it is to be understood that such conditions are capable of being varied by the skilled artisan who will appreciate the options available in the art for culturing cells.
  • isolated generally refers to a cell, which has been separated from its natural environment. This term includes gross physical separation from its natural environment, e.g., removal from the donor animal.
  • an isolated cell is not present in a tissue, i.e., the cell is separated or dissociated from the neighboring cells with which it is normally in contact.
  • cells are administered as a cell suspension.
  • the phrase "cell suspension” includes cells which are in contact with a medium and which have been dissociated, e.g., by subjecting a piece of tissue to gentle trituration.
  • peripheral blood mononuclear cell encompasses any blood cell having a round nucleus.
  • exemplary peripheral blood mononuclear cells include but are not limited to lymphocytes, monocytes, and macrophages.
  • This application provides for methods of detecting allogeneic therapeutic cells in a patient's blood sample after the cells have been administered to the patient.
  • the methods involve the steps of (a) providing a blood sample from a patient that has been treated with the allogeneic therapeutic cells; (b) analyzing the sample using an assay method to detect one or more markers positive for patient peripheral blood mononuclear cells (“PBMC”) (e.g. any blood cell having a round nucleus) and/or one or more markers positive for therapeutics cells; and (c) distinguishing between the human peripheral blood mononuclear cells and one or more markers positive for allogeneic therapeutic cells that are not expressed by the peripheral blood mononuclear cells.
  • PBMC peripheral blood mononuclear cells
  • the methods may further include the step of assaying for these markers.
  • the methods of the invention allow detection of such cells without being limited by the karyotype of the allogeneic therapeutic cell (i.e. XX vs. XY) and the gender of the patient.
  • the methods are not constrained by gender nor are they limited to only identifying male allogeneic therapeutic cells (XX) in a female patient.
  • the invention provides methods for detecting or identifying human umbilical cord tissue-derived cells in a blood sample.
  • the methods involve the steps of: (a) providing a blood sample from a patient that has been treated with human umbilical cord tissue-derived cells; (b) analyzing the sample using an assay method to detect one or more markers positive for human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells; and (c) distinguishing between the human peripheral blood mononuclear cells and the human umbilical cord tissue-derived cells.
  • an assay method to detect one or more markers positive for human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells
  • distinguishing between the human peripheral blood mononuclear cells and the human umbilical cord tissue-derived cells To able to distinguish, one also needs to assay for the unique marker profile to be able to select the distinguishing unique marker profile.
  • a further step of quantifying the amount of hUTC may also be employed.
  • the invention describes methods to distinguish and/or measure an hUTC cell therapy product following intravenous administration in humans.
  • the methods identify molecular markers that are expressed at substantially higher levels in hUTC as compared to cells normally present in blood, i.e., peripheral blood mononuclear cells ("PBMC"). Since the methods rely on a unique marker profile for the identification of hUTC, they allow detection of such cells without being limited by karyotype of the hUTC (XX vs. XY) and the gender of the patient. They are not constrained by gender nor are they limited to only male hUTC. Thus, these methods permit analysis of both male and female hUTC in both male and female subjects, in any combination.
  • PBMC peripheral blood mononuclear cells
  • the methods of the invention are suitable to distinguish allogeneic therapeutic cells in the blood of a patient from a variety of sources.
  • the methods of the invention are suitable for detecting a small number of hUTC in the presence of a large excess of a recipient's blood. This method is applicable for any member of the mammalian system and is not restricted to that derived from humans.
  • the methods of the invention may be used to distinguish allogeneic therapeutic cells such as e.g. hUTC from PBMC in a human.
  • the methods may be used to distinguish allogeneic therapeutic cells such as e.g. hUTC from PBMC in a non-human primate, mouse, rat, hamster, guinea pig, dog, or pig.
  • These methods help differentiate hUTC and other allogeneic therapeutic cells from the PBMC and may help monitor pharmacokinetics in subjects who receive a cell therapy product.
  • the methods of the invention are suitable for detection of allogeneic therapeutic cells (such as e.g. hUTC) in blood samples from patients having been treated with the cells.
  • the methods of the invention are suitable for detection of allogeneic therapeutic cells (such as e.g. hUTC) that may have been previously administered.
  • the methods disclosed herein may be suitable for the detection of any cell of therapeutic interest such as e.g. human cord blood-derived cells, placenta-derived cells, mesenchymal stem cells and mesenchymal stem cell-derived cells, hUTC, cardiomyocytes etc. or any targeted cells expressing a specific protein or proteins of interest.
  • Other suitable cells for use in these methods include liver cells, pancreatic islet cells, fibroblasts and insoluble collagenous bone matrix-derived (ICBM) cells.
  • the methods may also be suitable for detection of two or more kinds of allogeneic therapeutic cells (such as e.g. hUTC) in blood samples from patients having been treated with the cells.
  • the methods may be used to detect both human umbilical cord tissue-derived cells and placenta-derived cells in a blood sample.
  • the methods can be used to detect human umbilical cord tissue-derived cells and dermal fibroblasts in a blood sample.
  • the methods may also be used to detect human umbilical cord tissue-derived cells, placenta-derived cells, and dermal fibroblasts in a blood sample.
  • the methods of the invention include the step of identifying a distinguishing unique marker profile based on the presence of the therapeutic cell (a human umbilical cord tissue-derived cells) in a patient blood sample containing PBMC.
  • the expression of genes and presence of cell surface markers needs to be compared between the cells (e.g. human umbilical cord tissue-derived cells) and PBMC.
  • Multiple technologies including protein-based technologies such as ELISA and immune-histochemistry or nucleic acid based technologies such as in situ hybridization may be used to detect the presence of hUTC in blood.
  • PCR technology may be also used.
  • the expression level of various genes in PBMC and/or the allogeneic therapeutic cells may also be obtained from publically available sources.
  • the marker is a cell surface marker.
  • molecular markers on hUTC may be identified by comparing the expression profile of these cells to that of human peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • the unique marker profile may include one or more markers positive for the allogeneic cells, the presence of which is being assayed, and one or more markers positive for the peripheral blood mononuclear cells.
  • the marker profile when assaying for human umbilical cord tissue-derived cells, the marker profile may include one or more markers positive for hUTC and one or more markers positive for peripheral blood mononuclear cells.
  • the allogeneic cells, the presence of which is being assayed may be identified using only one or more unique markers for the allogeneic cells sufficient to distinguish these cells from the peripheral blood mononuclear cells.
  • the unique marker profile includes one or more of CD10, CD13, NRP1, CD45, LAMP1, DKK3, NRP1, or LAMB1.
  • the unique marker profile comprises at least one or more marker characteristic (e.g. positive) for the hUTC and at least one marker characteristic (e.g. positive) for PBMC (such as e.g. CD45).
  • the unique marker profile includes one or more unique markers that are positive for the hUTC and that are sufficient to distinguish hUTC from the peripheral blood mononuclear cells.
  • These one or more unique markers may include one or more of CD 10, CD13, NRP1, LAMP1, DKK3, NRP1, or LAMB1.
  • the choice of which marker to use may vary depending on which technique is used. For example, using a microarray analysis (such as e.g. Affymetrix GeneChip® HT HG-U133+ PM array), ANPEP (CD13), LAMP1 and LAMB1 may be insufficient to distinguish while one or more of the others markers may be sufficient to distinguish. Similarly, using RT-PCT, LAMP1 may not be suitable as part of the unique marker profile that distinguishes hUTC from PBMC, while one or more of the other markers may be sufficient.
  • the unique marker profile used for screening for hUTC by cell surface flow cytometry does not include DKK3 and LAMB1.
  • the unique marker profile for intracellular cytometry includes LAMP1 and DKK3.
  • Suitable markers for identifying hUTC in a PBMC sample using selected techniques are shown below.
  • the unique marker profile includes CD10 as the one or more markers positive hUTC and CD45 as the one or more markers positive for PBMC.
  • the unique marker profile includes CD10 and/or CD13 as the one or more markers positive hUTC and CD45 as the one or more markers positive for PBMC.
  • the unique marker profile for identifying hUTC in PBMC includes CD45 as the one or more markers positive for peripheral blood mononuclear cell and CD10 as one or more markers positive for human umbilical cord tissue-derived cells.
  • the maker profile to distinguish hUTC and PBMC comprises CD45, CD10, and CD13 using flow cytometry, wherein CD10 and CD13 are the one or more markers positive for hUTC and wherein CD45 is the one or more markers positive for PBMC.
  • Exemplary suitable markers for the identifying selected therapeutic cells in a PBMC sample are shown below: Type of cell Marker for cell type Marker for host PBMC Human umbilical cord-blood derived CD10, CD13, CD107a CD45 Placenta derived Renin Heat shock 27kd Mesenchymal stem cell derived IL26, a-glucosidase CD45 Liver FOXa2, HNFal CD45 Pancreatic islet cells G6PC2, INSULIN CD45 Fibroblast SDF-1, GCP-2 CD45 Human umbilical cord-blood derived and placenta-derived cells and dermal fibroblasts IL8, GCP-2 CD45 Human umbilical cord-blood derived and placenta-derived cells IL6, MCP-1 CD45 Insoluble collagenous bone matrix (ICBM) Integrin a-10; cardiac ankyrin repeat protein CD45 Cardiomyocytes FoxA2,GATA4, MIR20 CD45
  • the unique marker profile is obtainable using the following procedures as well the methodology described in the examples.
  • the procedures describe below may encompass both obtaining the unique marker profile and verifying its accuracy. While the step of verification may encompass analysis of a sample, the step may be necessary to determine the accuracy and testing limits of the specific procedure by which the unique marker gene or protein will be detected. In this embodiment, flow cytometry may be used to identify the markers.
  • the gene expression profile of hUTC sourced from multiple sources, was generated using an Affymetrix GeneChip® HT HG-U133+ PM.
  • the gene expression profiles of human and rat PBMC and other types of blood cells were obtained from publicly available databases. From the results of the gene expression profiles, a set of markers that are highly expressed in hUTC and a set of markers that are highly expressed in human PBMC can be identified. Sixty-one (61) genes were thus identified, a subset of which were studied in more detail. From this list of 61 genes, CD45 was identified as a positive marker for hPBMC and CD10 and CD13 as two positive markers for hUTC.
  • Verifying the testing methodology for the marker profile Since the number of hUTC in a sample of a subject's blood is anticipated to be low, it was determined whether a small number of hUTC in the presence of a large excess of human PBMC using the above-identified markers could be detected and quantified. For this purpose, known quantities of hUTC were spiked into approximately one million PBMC in 1 ml of human serum. Subsequently, an aliquot of each sample was analyzed by flow cytometry for CD45 as a positive marker for hPBMC and CD10 or CD13 as positive markers for hUTC. The cells were harvested by centrifugation, washed once with PBS and then fixed and permeabilized.
  • the unique marker profile may be provided as part of a kit containing the materials for testing for the unique marker profile.
  • the step of identifying the unique marker profile may also include the step of verifying its correctness and possible validation of the unique marker profile in compliance with relevant regulations.
  • the step of obtaining a patient sample includes the step of taking a blood sample from the patient.
  • the patient may be a human, non-human primate, mouse, rat, hamster, guinea pig, dog, or pig.
  • the blood sample may be further purified or concentrated to isolate the cells.
  • the plasma may be removed from the blood.
  • the plasma may be removed using standard techniques in the art including centrifugation.
  • the patient sample (e.g. fraction) used for further analysis comprises the allogeneic therapeutic cells of interest and the peripheral blood mononuclear cells from the patient.
  • the patient sample comprises a human umbilical cord tissue-derived/peripheral blood mononuclear cell fraction.
  • the step of obtaining a patient sample further includes isolating the human umbilical cord tissue-derived/peripheral blood mononuclear cell fraction from the blood sample.
  • This embodiment may further include the step of removing plasma.
  • the step of obtaining a patient sample includes use of Ficoll extraction.
  • the step of analysis comprises taking the patient blood sample and testing it for the presence of the distinguishing unique marker profile for allogeneic therapeutic cells (e.g. hUTC).
  • the step of analysis may also include determining the presence of PBMC based on a unique marker profile as determined above.
  • the step of analyzing the patient sample includes use of an assay method to test for one or more markers positive for the allogeneic therapeutic cells and one or more markers positive for peripheral blood mononuclear cells.
  • the step of analysis includes detecting the presence of cells based on the unique marker profile assayed above.
  • the step of analyzing the patient sample includes use of an assay method to test for one or more unique markers positive for the allogeneic therapeutic cell sufficient to distinguish these cells from the peripheral blood mononuclear cells.
  • the step of analysis includes detecting one or more markers positive for peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells in the patient blood sample. In another embodiment, the step of analysis includes detecting one or more unique markets positive for hUTC sufficient to distinguish these cells from PBMC in the patient blood sample.
  • the patient blood sample may be a human umbilical cord tissue-derived/peripheral blood mononuclear cell fraction.
  • the step of analysis may be carried out using a variety of standard laboratory techniques.
  • the step of analyzing may utilize flow cytometry, ELISA, immunohistochemistry, nucleic acid detection, PCR, or combinations thereof.
  • telomeres may be used to detect the presence of hUTC in blood.
  • PCR technology may be used to facilitate the relative quantification of hUTC in a blood sample.
  • the methods of the invention are used to distinguish human hUTC from human PBMC.
  • the unique marker profile comprises one or more of CD10, CD13, NRP1, CD45, LAMP1, DKK3, NRP1, or LAMB1 and the step of analyzing may utilize flow cytometry, ELISA, immunohistochemistry, nucleic acid detection, PCR, or combinations thereof.
  • step of analysis includes flow cytometry to test for CD 10 and/or CD 13 as a positive marker for human umbilical cord tissue-derived cells and CD45 as a positive marker for human peripheral blood mononuclear cells.
  • the methods of the invention are used to distinguish hUTC from human PBMC based on the presence of one or more unique positive markers (such as e.g. CD10, CD13, NRP1, LAMP1, DKK3, NRP1, or LAMB1) sufficient to distinguish these cells from the PBMC.
  • one or more unique positive markers such as e.g. CD10, CD13, NRP1, LAMP1, DKK3, NRP1, or LAMB1
  • the methods of the invention may further include one or more enrichment steps.
  • enrichment refers to the process of substantially increasing the ratio of target bioentities (e.g., allogeneic therapeutic cells (such as e.g. hUTC)) to non-target materials (e.g. PBMC) in the processed analytical sample compared to the ratio in the original biological sample.
  • target bioentities e.g., allogeneic therapeutic cells (such as e.g. hUTC)
  • non-target materials e.g. PBMC
  • an enrichment step may be necessary prior to analyzing the patient sample.
  • an enrichment step may be necessary where there are so few allogeneic therapeutic cells (e.g. hUTC) present relative to the number of blood cells or that the amount of allogeneic therapeutic cells falls below the limits of detection of the laboratory technique.
  • the methods of the invention can identify and quantify allogeneic therapeutic cells, even if present in very low concentration in the patient's blood and provide a mechanism to facilitate quantification of cells present in any given sample.
  • a variety of standard techniques may be used for the one or more enrichment steps. These techniques include, but are not limited to, the use of selective medium for enrichment and depletion of specific cell types, selective adhesions method, physical and biological methods of cell separation, density gradient electrophoresis, centrifugation and the like. Exemplary enrichment techniques are immunoaffinity column, immunoaffinity beads, centrifugation through a sucrose or Ficoll gradient.
  • Anti-CD45 antibody conjugated to magnetic beads may be used for this purpose (see discussion below).
  • the enrichment step may include cell separation with magnetic particles/beads.
  • Magnetic particles are well known in the art, as is their use in immune and other bio-specific affinity reactions. Generally, any material that facilitates magnetic or gravitational separation may be employed for this purpose. Exemplary enrichment procedures using magnetic beads are described in U.S. Patent No. 7,863,012 and U.S. Published Application No. 2011/0147180 .
  • Magnetic particles can be classified based on size: large (1.5 to about 50 microns); small (about 0.7-1.5 microns); or colloidal ( ⁇ 200 nm), which are also referred to as nanoparticles.
  • Small magnetic particles of the type described above are quite useful in analyses involving bio-specific affinity reactions, as they are conveniently coated with biofunctional polymers (e.g. , proteins), provide very high surface areas, and give reasonable reaction kinetics.
  • biofunctional polymers e.g. , proteins
  • Magnetic particles ranging from about 0.7-1.5 microns have been described in the patent literature, including, by way of example, U.S. Pat. Nos. 3,970,518 ; 4,018,886 ; 4,230,685 ; 4,267,234 ; 4,452,773 ; 4,554,088 ; and 4,659,678 . Certain of these particles are disclosed to be useful solid supports for immunological reagents.
  • the efficiency with which magnetic separations can be done and the recovery and purity of magnetically labeled cells will depend on many factors. These include: number of cells being separated, receptor or epitope density of such cells, magnetic load per cell, non-specific binding (NSB) of the magnetic material, carry-over of entrapped non-target cells, technique employed, nature of the vessel, nature of the vessel surface, viscosity of the medium, and magnetic separation device employed. If the level of non-specific binding of a system is substantially constant, as is usually the case, then as the target population decreases so will the purity.
  • a system with 0.8% NSB that recovers 80% of a population, which is at 0.25% in the original mixture will have a purity of 25%.
  • the initial population was at 0.01% (one target cell in 10 6 bystander cells)
  • the NSB were 0.001%
  • the purity would be 8%.
  • Extremely low non-specific binding is required or advantageous to facilitate detection and analysis of rare cells, such as epithelial derived tumor cells present in the circulation.
  • a targeted cell such as e.g. hUTC
  • labeling and recovery will markedly depend on the nature of magnetic particle employed. For example, when cells are incubated with large magnetic particles, such as Dynal® magnetic beads (Invitrogen), cells are labeled through collisions created by mixing of the system, as the beads are too large to diffuse effectively. Thus, if a cell were present in a population at a frequency of 1 cell per ml of blood or even less, then the probability of labeling target cells will be related to the number of magnetic particles added to the system and the length of time of mixing.
  • the magnetic particles for use in carrying out the enrichment step behave as colloids.
  • Such particles are characterized by their sub-micron particle size, which is generally less than about 200 nm (0.20 microns), and their stability to gravitational separation from solution for extended periods. In addition to the many other advantages, this size range makes them essentially invisible to analytical techniques commonly applied to cell analysis. Particles within the range of about 90-150 nm and having between about 70-90% magnetic mass are contemplated for use in the present invention.
  • Suitable magnetic particles are composed of a crystalline core of superparamagnetic material surrounded by molecules, which are bonded, e.g., physically absorbed, or covalently attached to the magnetic core and which confer stabilizing colloidal properties.
  • the coating material should preferably be applied in an amount effective to prevent non-specific interactions between biological macromolecules found in the sample and the magnetic cores.
  • biological macromolecules may include carbohydrates such as sialic acid residues on the surface of non-target cells, lectins, glycoproteins, and other membrane components.
  • the material should contain as much magnetic mass per nanoparticle as possible.
  • the size of the magnetic crystals comprising the core is sufficiently small that they do not contain a complete magnetic domain.
  • the size of the nanoparticles is sufficiently small such that their Brownian energy exceeds their magnetic moment. Consequently, North Pole, South Pole alignment and subsequent mutual attraction/repulsion of these colloidal magnetic particles does not appear to occur even in moderately strong magnetic fields, contributing to their solution stability.
  • magnetic particles should be separable in high magnetic gradient external field separators. That characteristic facilitates sample handling and provides economic advantages over the more complicated internal gradient columns loaded with ferromagnetic beads or steel wool.
  • Magnetic particles having the above-described properties can be prepared by modification of base materials described in U.S. Pat. Nos. 4,795,698 ; 5,597,531 , and 5,698,271 .
  • small nanoparticles (30-70 nm) will diffuse more readily, they will preferentially label cells compared with their larger counterparts.
  • very high gradients such as in internal gradient columns
  • the performance of these materials regardless of size, makes little difference.
  • external gradients, or gradients of lesser magnitude than can be generated on micro bead or steel wool columns the occupancy of small nanoparticles on cells has a significant effect. This was conclusively shown to be the case by fractionating DC nanoparticles and studying the effects on recovery. Based on these studies and other optimization experiments, means for fractionating nanoparticles magnetically or on columns was established where base coated magnetic particles could be prepared that were devoid of excessively small or large nanoparticles.
  • base coated particles of mean diameter 100 nm can be produced which contain at best trace amounts of material smaller than 80 nm or over 130 nm.
  • material of about 120 nm can be made with no appreciable material smaller than 90-95 nm and over 160 nm.
  • Such materials performed optimally with regard to recovery and could be made sub-optimal by the inclusion of 60-70 nm nanoparticles.
  • One preferred particle size range for use in practicing this invention is 90-150 nm for base coated magnetic particles, e.g., BSA-coated magnetite.
  • high gradient magnetic separation with an external field device employing highly magnetic, low non-specific binding, colloidal magnetic particles is the method of choice for separating a cell subset of interest from a mixed population of eukaryotic cells (such as e.g. allogeneic therapeutic cells (e.g. hUTC) in peripheral blood mononuclear cells), particularly if the subset of interest comprises but a small fraction of the entire population.
  • eukaryotic cells such as e.g. allogeneic therapeutic cells (e.g. hUTC) in peripheral blood mononuclear cells
  • the magnetic particles must be specific for epitopes that are not present on the allogeneic therapeutic cells (e.g. hUTC).
  • An enrichment step by use of magnetic beads includes using magnetic nanoparticles (such as those described above) which are labeled with a monoclonal antibody.
  • These monoclonal antibodies may be antibodies identifying peripheral blood mononuclear cells but not allogeneic therapeutic cells.
  • the monoclonal antibody attached to the nanoparticles bind to the peripheral blood mononuclear cells which may then be separated using magnetic means. Typically, such separation is achieved via the use of a high magnetic gradient external field separators.
  • An exemplary suitable technique to separate by magnetic means is described in U.S. Pat. No. 6,365,362 .
  • an Anti-CD45 antibody conjugated to a magnetic particle is used.
  • the enrichment step includes use of Anti-CD4 and anti-CD8 antibodies, to remove T-cells.
  • the enrichment step includes monoclonal antibodies identifying the allogeneic therapeutic cells.
  • the allogeneic therapeutic cells are hUTC and the antibodies include one or more of anti-CD 10 or anti-CD13 antibodies.
  • the methods of the invention include use of a Cell Search System (Veridex, LLC (Raritan, NJ)), which has been optimized for the detection of hUTC and PBMC.
  • a Cell Search System (Veridex, LLC (Raritan, NJ)
  • the methods of the invention may also include the step of preserving a sufficient patient sample to run an additional analysis. This may be particularly useful where initial analysis might not be able to detect the allogeneic therapeutic cells (such as e.g. hUTC) due to allogeneic therapeutic cells being present in an amount below the detection limit of the assay technique used.
  • the method further includes an additional analysis of a patient sample if no allogeneic therapeutic cells are initially detected (such as e.g. hUTC). This additional analysis also includes one or more enrichment steps such as those discussed above.
  • the methods of the invention further comprise determining the presence of allogeneic therapeutic cells in a sample of peripheral blood mononuclear cells.
  • the step of determining the presence of said cells includes comparing the results of the analysis of the sample to the unique marker, which was assayed for above, wherein the presence of unique markers for allogeneic therapeutic cells indicates the presence of the allogeneic therapeutic cells in the sample.
  • the step of determining may also include comparing the result to a unique marker for the PBMC as identified above.
  • the determination step includes comparing the results for the one or more markers positive for the allogeneic therapeutic cell (hUTC) and the one or more markers negative for PBMC to the marker profile above.
  • the method of the invention comprises determining the presence of human umbilical cord tissue-derived in a sample of peripheral blood mononuclear cells (such as e.g. human or rodent).
  • the determination step includes comparing the results of the analysis of the sample to the unique marker, which was assayed for above, wherein the presence of unique markers for the allogeneic therapeutic cells indicates the presence of the hUTC cells in the sample.
  • the determination step may also include comparing the result to a unique marker for the PBMC as identified above.
  • the determination step may include one or more markers positive for the hUTC and one or more markers positive for PBMC as discussed above.
  • the determination step also includes distinguishing between the human peripheral blood mononuclear cells and human umbilical cord tissue-derived cells based on the detection of one or more markers positive for human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells.
  • the determination step may also include differentiating between human umbilical cord tissue-derived cells administered to the patient and human peripheral blood mononuclear cells from the patient.
  • the step of determining the presence of the cells may also include quantifying the cells.
  • the determination step for the presence of hUTC includes quantifying the number of hUTC in the sample.
  • the step of determining the presence may further include selecting one or more human umbilical cord tissue-derived cells based on the presence of the unique marker profile (such as e.g. one or more markers positive for hUTC).
  • the determination step may also include detecting the presence of the human peripheral blood mononuclear cells and human umbilical cord tissue-derived cells based on the detection of CD45 as a marker positive for human peripheral blood mononuclear cells and CD10 or CD13 as a marker positive for human umbilical cord tissue-derived cells.
  • the methods described herein may be suitable to distinguish allogeneic therapeutic cells from rat PBMC, which may be particularly useful for monitoring in rat disease models.
  • the present invention describes a method for the identification of hUTC in blood.
  • the method involves the following steps: (a) providing a blood sample from a patient that has been treated with human umbilical cord tissue-derived cells; (b) analyzing the sample using an assay method to detect one or more markers positive for human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells; and (c) distinguishing between the human peripheral blood mononuclear cells and one or more markers positive for human umbilical cord tissue-derived cells.
  • a further step of quantifying the amount of hUTC may also be employed. In one embodiment, one or more of the markers shown in Example 3 are used.
  • the methods of the invention are suitable for identifying about 1,700 or more hUTC/ml in the presence of 1 million human PBMC using flow cytometry without relying on an enrichment step.
  • Another embodiment disclosed herein is a kit for testing for the presence of allogeneic therapeutic cells such as hUTC in a blood sample using the disclosed methods.
  • kits may comprise the distinguishing unique marker profile and suitable components for identifying the distinguishing unique marker profile according to one or more analytical procedures.
  • a kit suitable for distinguishing a unique marker profile by using RT-PCR would include PCR primers suitable for amplifying the unique marker profile genes.
  • a kit suitable for distinguishing a unique marker profile by an antibody-linked assay would include the antibodies that bind to the markers.
  • the kit is designed to detect hUTC in a population of PBMC.
  • the kit comprises the unique marker profile CD10, CD13 and CD45 and other suitable components for identifying CD10, CD13 and CD45 using one or more of flow cytometry, ELISA, immunohistochemistry, nucleic acid detection, PCR or combinations thereof.
  • Yet another embodiment is a system for detecting allogeneic therapeutic cells (such as e.g. hUTC) in a population of PBMC.
  • the system includes materials necessary for carrying out the methods disclosed herein.
  • the methods may be used to distinguish therapeutic (e.g. progenitor/stem cells) from host cells
  • the cells are human umbilical cord tissue-derived-cells ("hUTC” or "UTC”).
  • hUTC human umbilical cord tissue-derived-cells
  • Useful human umbilical cord tissue-derived cells are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, and have the potential to differentiate.
  • the UTC and UTC populations suitable for identification by the methods of the invention are described in detail in detailed herein below as well as U.S. Patent Nos. 7,510,873 ; 7,524,489 ; and U.S. Pub. App. No. 2005/0058631 .
  • a mammalian umbilical cord is recovered upon or shortly after termination of either a full-term or a pre-term pregnancy, e.g., after expulsion of after birth.
  • the postpartum tissue may be transported from the birth site to a laboratory in a sterile container such as a flask, beaker, culture dish, or bag.
  • the container may have a solution or medium, including but not limited to a salt solution, such as Dulbecco's Modified Eagle's Medium (DMEM) (also known as Dulbecco's Minimal Essential Medium) or phosphate buffered saline (PBS), or any solution used for transportation of organs used for transplantation, such as University of Wisconsin solution or perfluorochemical solution.
  • DMEM Dulbecco's Modified Eagle's Medium
  • PBS phosphate buffered saline
  • antibiotic and/or antimycotic agents such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin, may be added to the medium or buffer.
  • the postpartum tissue may be rinsed with an anticoagulant solution such as heparin-containing solution. It is preferable to keep the tissue at about 4-10 °C prior to extraction of UTC. It is even more preferable that the tissue not be frozen prior to extraction of UTC.
  • Isolation of UTC preferably occurs in an aseptic environment.
  • the umbilical cord may be separated from the placenta by means known in the art.
  • Blood and debris are preferably removed from the postpartum tissue prior to isolation of UTC.
  • the postpartum tissue may be washed with buffer solution, including but not limited to phosphate buffered saline.
  • the wash buffer also may comprise one or more antimycotic and/or antibiotic agents, including but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.
  • Postpartum tissue comprising an umbilical cord or a fragment or section thereof is disaggregated by mechanical force (mincing or shear forces).
  • the isolation procedure also utilizes an enzymatic digestion process.
  • Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture. Digestion enzymes range from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), and are available commercially.
  • a non-exhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases.
  • enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities.
  • collagenases are known to be useful for isolating various cells from tissues.
  • Deoxyribonucleases can digest single-stranded DNA and can minimize cell clumping during isolation.
  • Preferred methods involve enzymatic treatment with e.g. collagenase and dispase, or collagenase, dispase, and hyaluronidase.
  • a mixture of collagenase and the neutral protease dispase are used in the dissociating step. More specific embodiments employ digestion in the presence of at least one collagenase from Clostridium histolyticum, and either of the protease activities, dispase, and thermolysin. Still other embodiments employ digestion with both collagenase and dispase enzyme activities. Also utilized are methods that include digestion with a hyaluronidase activity in addition to collagenase and dispase activities. The skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources.
  • the enzyme blends for tissue disassociation sold under the trade name LIBERASE (Roche, Indianapolis, Ind.) are suitable for use in the instant methods.
  • Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources.
  • the skilled artisan is also well-equipped to assess new or additional enzymes or enzyme combinations for their utility in isolating the cells.
  • Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer.
  • the tissue is incubated at 37°C during the enzyme treatment of the dissociation step.
  • postpartum tissue is separated into sections comprising various aspects of the tissue, such as neonatal, neonatal/maternal, and maternal aspects of the placenta, for instance.
  • the separated sections then are dissociated by mechanical and/or enzymatic dissociation according to the methods described herein.
  • Cells of neonatal or maternal lineage may be identified by any means known in the art, e.g. by karyotype analysis or in situ hybridization for a Y chromosome.
  • Isolated cells or umbilical cord tissue from which a UTC is derived may be used to initiate, or seed, cell cultures. Isolated cells are transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (native, denatured or cross-linked), gelatin, fibronectin, and other extracellular matrix proteins.
  • extracellular matrix or ligands such as laminin, collagen (native, denatured or cross-linked), gelatin, fibronectin, and other extracellular matrix proteins.
  • a UTC may be cultured in any culture medium capable of sustaining growth of the cell such as, but not limited to, DMEM (high or low glucose), advanced DMEM, DMEM/MCDB 201, Eagle's basal medium, Ham's F10 medium (F10), Ham's F-12 medium (F12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM), DMEM/F12, RPMI 1640, and serum/media free medium sold under the trade name CELL-GRO-FREE (Mediatch, Inc., Herndon, Va.).
  • the culture medium may be supplemented with one or more components including, e.g., fetal bovine serum (FBS), preferably about 2-15% (v/v); equine serum (ES); human serum (HS); beta-mercaptoethanol (BME or 2-ME), preferably about 0.001% (v/v); one or more growth factors, e.g., platelet-derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), leukocyte inhibitory factor (LIF) and erythropoietin (EPO); amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contamination, such as penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination.
  • the culture medium may comprise Growth Medium as defined in the Examples below.
  • the cells are seeded in culture vessels at a density to allow cell growth.
  • the cells are cultured at about 0 to about 5% by volume CO 2 in air.
  • the cells are cultured at about 2 to about 25% O 2 in air, preferably about 5 to about 20% O 2 in air.
  • the cells preferably are cultured at a temperature of about 25 to about 40° C. and more preferably are cultured at 37° C.
  • the cells are preferably cultured in an incubator.
  • the medium in the culture vessel can be static or agitated, e.g., using a bioreactor.
  • the UTC is preferably grown under low oxidative stress (e.g., with addition of glutathione, Vitamin C, Catalase, Vitamin E, N-Acetylcysteine).
  • Low oxidative stress refers to conditions of no or minimal free radical damage to the cultured cells.
  • the UTC After culturing the isolated cells or tissue fragments for a sufficient period, a UTC will have grown out, either because of migration from the postpartum tissue or cell division, or both.
  • the UTC is passaged, or removed to a separate culture vessel containing fresh medium of the same or a different type as that used initially, where the population of cells can be mitotically expanded.
  • the cells may be used at any point between passage 0 and senescence.
  • the cells preferably are passaged between about 3 and about 25 times, more preferably are passaged about 4 to about 12 times, and preferably are passaged 10 or 11 times. Cloning and/or subcloning may be performed to confirm that a clonal population of cells has been isolated.
  • the different cell types present in postpartum tissue are fractionated into subpopulations from which the UTC can be isolated. Fractionation or selection may be accomplished using standard techniques for cell separation including, but not limited to, enzymatic treatment to dissociate postpartum tissue into its component cells, followed by cloning and selection of specific cell types, including but not limited to selection based on morphological and/or biochemical markers; selective growth of desired cells (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population such as, e.g., with soybean agglutinin; freeze-thaw procedures; differential adherence properties of the cells in the mixed population; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent distribution; electrophoresis; and fluorescence activated cell sorting (FACS).
  • FACS fluorescence activated cell sorting
  • the culture medium is changed as necessary, e.g., by carefully aspirating the medium from the dish, e.g., with a pipette, and replenishing with fresh medium. Incubation is continued until a sufficient number or density of cells accumulates in the dish.
  • the original explanted tissue sections may be removed and the remaining cells trypsinized using standard techniques or using a cell scraper. After trypsinization, the cells are collected, removed to fresh medium, and incubated as above. In some embodiments, the medium is changed at least once at approximately 24 hours post-trypsinization to remove any floating cells. The cells remaining in culture are considered to be UTC.
  • the UTC may be cryopreserved. Accordingly, UTC for autologous transfer (for either the mother or child) may be derived from appropriate postpartum tissues following the birth of a child, then cryopreserved so as to be available in the event they are later needed for transplantation.
  • hUTC may be distinguished from PBMC based on the presence of e.g. CD45, CD13, and CD10 and other markers discussed above and in the examples below, hUTC possess a variety of other unique characteristics.
  • the UTC may be characterized, e.g., by growth characteristics (e.g., population doubling capability, doubling time, passages to senescence), karyotype analysis (e.g., normal karyotype; maternal or neonatal lineage), flow cytometry (e.g., FACS analysis), immunohistochemistry and/or immunocytochemistry (e.g., for detection of epitopes), gene expression profiling (e.g., gene chip arrays; polymerase chain reaction (e.g., reverse transcriptase PCR, real time PCR, and conventional PCR)), protein arrays, protein secretion (e.g., by plasma clotting assay or analysis of PDC-conditioned medium, e.g., by Enzyme Linked ImmunoSorbent Assay (ELISA)), mixed lymphocyte reaction (e.g. , as measure of stimulation of PBMCs), and/or other methods known in the art.
  • growth characteristics e.g., population doubling capability, doubling time,
  • UTC derived from umbilicus tissue were deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110) on June 10, 2004, and assigned ATCC Accession Numbers as follows: (1) strain designation UMB 022803 (P7) was assigned Accession No. PTA-6067; and (2) strain designation UMB 022803 (P17) was assigned Accession No. PTA-6068.
  • the UTC possesses one or more of the following growth features: (1) they require L-valine for growth in culture; (2) they are capable of growth in atmospheres containing oxygen from about 5% to at least about 20%; (3) they have the potential for at least about 40 doublings in culture before reaching senescence; and (4) they attach and expand on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin or fibronectin.
  • the UTC possesses a normal karyotype, which is maintained as the cells are passaged.
  • Methods for karyotyping are available and known to those of skill in the art.
  • the UTC may be characterized by production of certain proteins, including: (1) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; and (2) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C cell surface markers, as detected by flow cytometry.
  • the UTC may be characterized by lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR, DP, DQ cell surface markers, as detected by flow cytometry.
  • Particularly preferred are cells that produce at least two of tissue factor, vimentin, and alpha-smooth muscle actin. Also preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
  • the UTC may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for a gene encoding at least one of: interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
  • interleukin 8 reticulon 1
  • chemokine (C-X-C motif) ligand 1 melonoma growth stimulating activity, alpha
  • chemokine (C-X-C motif) ligand 6 granulocyte chemotactic protein 2
  • chemokine (C-X-C motif) ligand 3 and tumor necrosis
  • the UTC may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is reduced for a gene encoding at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1;
  • the UTC may be characterized when cultured by secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1b, 1309, MDC RANTES, and TIMP1.
  • the UTC may be characterized when cultured by lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1A, and VEGF.
  • the UTC are derived from umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, require L-valine for growth, can grow in at least about 5% oxygen, and comprise at least one of the following characteristics: potential for at least about 40 doublings in culture; attachment and expansion on a coated or uncoated tissue culture vessel that comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin, or fibronectin; production of vimentin and alpha-smooth muscle actin; production of CD10, CD13, CD44, CD73, and CD90; and, expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is increased for a gene encoding interleukin 8 and reticulon 1. In some embodiments, such UTC does not produce CD45 and CD117.
  • the cell comprises two or more of the above-listed growth, protein/surface marker production, gene expression, or substance-secretion characteristics. More preferred is a cell comprising three, four, five, or more of the characteristics. Still more preferred is a UTC comprising six, seven, eight, or more of the characteristics. More preferred is a cell comprising all of above characteristics.
  • the UTC are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, lack the production of CD117 or CD45, express CD10 and CD13, and do not express hTERT or telomerase.
  • These UTC optionally express oxidized low density lipoprotein receptor 1, reticulon, chemokine receptor ligand 3, and/or granulocyte chemotactic protein; and/or do not express CD31 or CD34; and/or express, relative to a human fibroblast, mesenchymal stem cell, or iliac crest bone marrow cell, increased levels of interleukin 8 or reticulon 1; and/or express CD44, CD73, and CD90.
  • the UTC are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, express CD10, CD13, CD90, and HLA-ABC, and do not express CD34, CD45, CD117, and HLA-DR.
  • these cells also do not express hTERT or telomerase.
  • the cells also express CD44, and CD43.
  • the cells also do not express CD31.
  • UTC optionally: (i) express oxidized low density lipoprotein receptor 1, reticulon, chemokine receptor ligand 3, and/or granulocyte chemotactic protein; and/or (ii) express, relative to a human fibroblast, mesenchymal stem cell, or iliac crest bone marrow cell, increased levels of interleukin 8 or reticulon 1.
  • the UTC are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, and have the following characteristics: (1) express CD10, CD13, CD44, CD90, and HLA-ABC; (2) do not express CD31, CD34, CD45, HLA-DR and CD117, and (3) do not express hTERT or telomerase.
  • the UTC are isolated from human umbilical cord tissue substantially free of blood, are capable of self-renewal and expansion in culture, have the potential to differentiate, and have the following characteristics: (1) express CD10, CD13, CD44, CD90, and HLA-ABC; (2) do not express CD31, CD34, CD45, HLA-DR and CD117; (3) do not express hTERT or telomerase; (4) express oxidized low density lipoprotein receptor 1, reticulon, chemokine receptor ligand 3, and/or granulocyte chemotactic protein; and (5) express, relative to a human fibroblast, mesenchymal stem cell, or iliac crest bone marrow cell, increased levels of interleukin 8 or reticulon 1.
  • the hUTC are provided as a population of cells, which may be homogenous.
  • the cell population may be heterogeneous.
  • a heterogeneous cell population may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% UTC.
  • the heterogeneous cell populations may further comprise stem cells or other progenitor cells, such as myoblasts or other muscle progenitor cells, hemangioblasts, or blood vessel precursor cells; or it may further comprise fully differentiated skeletal muscle cells, smooth muscle cells, pericytes, or blood vessel endothelial cells.
  • the population is substantially
  • the hUTC which may be identified using the screening methods may be isolated and characterized according to the disclosure of U.S. Patent Nos. 7,510,873 ; 7,524,489 ; and U.S. Pub. App. No. 2005/0058631 , which relate to the description, isolation and characterization of hUTC.
  • enrichment procedures using magnetic beads are described in U.S. Patent No. 7,863,012 and U.S. Published Application No. 2011/0147180 , which relate to enrichment procedures and magnetic beads.
  • Human umbilical cord tissue-derived-cells were isolated from four donors and propagated in growth medium supplemented with 15% fetal bovine serum as described above in U.S. Patent Nos. 7,510,873 ; 7,524,489 ; and U.S. Pub. App. No. 2005/0058631 and in the Examples below.
  • Early passage cultures were cryopreserved to generate development and working cell banks, termed DCB and WCB, respectively.
  • Live cultures of hUTC were maintained by using one of the following two methods: 1) as adherent cultures in tissue culture flasks; and 2) as suspension cultures in spinner flasks and stirred tank bioreactors. Cells were first seeded onto microcarriers for the latter method.
  • cells were cultured in spinner flasks, starting from an early passage cell line population doubling PDL 5 and ending with PDL44.
  • a typical culture was initiated by seeding fresh growth medium with an inoculum of about 5 x 10 5 cells/ml and left undisturbed for 4 days, at which time a peak viable cell density (VCD of about 3-4 x 10 6 cells/ml) was achieved. Aliquots of cultures were harvested periodically, for a total of thirty-two (32) samples. Table 1-1 below lists the samples in the study.
  • Figure 1 shows the population doubling level (PDL) and the region of the growth curve when a particular sample was harvested.
  • An Affymetrix GeneChip® HT HG-U133+ PM array was used for microarray data generation.
  • the data was analyzed in two stages.
  • the microarray data generated from the temperature excursion study (Study 2, Table 1-2) to compare the expression profile of hUTC with that of human PBMC.
  • the expression level of various genes in PBMC was obtained from public sources, including, the National Center for Biotechnology (NCBI) database, administered by the National Institutes of Health.
  • the microarray data generated from Study 1 and archived data from eight (8) samples generated in Study 3 were used.
  • the expression profile of hUTC was compared with that of (1) human PBMC and (2) rat PBMC.
  • the expression profile of human PBMC was obtained from healthy human subjects (NCBI database study GSE14642), while the expression profile of rat PBMC was obtained from NCBI database studies GSE11083 and GSE19537. Expression profiles of subtypes of human blood cells (B cell, NK cell, dendritic cell, lymphoid T cell, myeloid monocyte and neutrophils) were also obtained from NCBI databases (study GSE22886 and study GSE3982). Additionally, mining the Gene Ontology and Entrez databases identified 3976 human plasma membrane protein genes (GO:0005886), 355 cell surface protein genes (GO:0009986) and 349 CD antigen genes. A subset of 2614 human and rat plasma membrane protein genes and 288 cell surface protein genes were matched to genes on the human and rat microarrays and 315 CD antigen genes and were matched to human microarrays were included for this study.
  • genes that were identified in all three sets of samples are underlined. Genes that were examined in more detail are bolded (they include DKK3, LAMB1, ANPEP (CD13), LAMP1 (CD107a), PTPRC (CD45), MME (CD10) and NRP1).
  • Table 1-4 List of genes whose expression levels were significantly different between hUTC and that of human PBMC in Study 2 samples Gene Symbol Entrez Gene Gene Title KRT19 3880 Keratin 19 DKK3 27122 Dickkopf homolog 3 ( Xenopus laevis ) GJA1 2697 Gap junction protein, alpha 1, 43kDa LOX 4015 Lysyl oxidase FBN1 2200 Fibrillin 1 COL3A1 1281 Collagen, type III, alpha 1 TPM2 7169 Tropomyosin 2 (beta) COL1A1 1277 Collagen, type I, alpha 1 CYR61 3491 Cysteine-rich, angiogenic inducer, 61 DKK1 22943 Dickkopf homolog 1 ( Xenopus laevis ) CTGF 1490 Connective tissue growth factor DCBLD2 131566 Discoidin, CUB and LCCL domain containing ITGB5 3693 Integrin, beta 5 COL5A1 1289 Collagen, type V, al
  • Table 1-5 shows the list of genes (generated in Study 1 and Study 2), whose expression levels were significantly different between hUTC and that of human PBMC.
  • underlining indicates genes that were identified in all three sets of samples, i.e., Study 1, 2, and 3 (they include LAMB1, DKK3 and CAP2). Genes that were examined in more detail are bolded (they include ANPEP (CD13), LAMP1 (CD107a), PTPRC (CD45), MME (CD10) and NRP1).
  • Table 1-5 List of genes whose expression levels were significantly different between hUTC and that of human PBMC in samples generated for Study 2 and 3 Gene Symbol Entrees Gene Gene Title LAMB1 3912 laminin beta 1 LUM 4060 Lumican WWTR1 25937 WW domain containing transcription regulator DKK3 27122 dickkopf homolog 3 ( Xenopus laevis ) NR2F2 7026 nuclear receptor subfamily 2, group F, member CAP2 10486 CAP, adenylate cyclase-associated protein, 2 ANPEP (CD13) 290 alanyl (membrane) aminopeptidase LAMP1 (CD107a) 3916 lysosomal-associated membrane protein 1 PTPRC (CD45) 5788 protein tyrosine phosphatase, receptor type, C MME (CD10) 4311 matrix metallo protease NRP1 8829 neuropilin 1
  • NRP1, DKK3, LAMP1, LAMB1 and MME are cell surface markers while the expressed proteins, PTPRC (CD45) and ANPEP (CD13) are localized intracellularly.
  • PTPRC CD45
  • MME CD10
  • ANPEP CD13
  • RNA from each hUTC preparation was isolated from which cDNA was then prepared. Fluorescent probes specific for each gene were then used to perform the RT-PCR reaction using the cDNA as the template.
  • Figure 3 shows the results of RT-PCR for MME (CD10), ANPEP (CD13), PTPRC (CD45), DKK3, LAMB1, NRP1, GAPPD, and HPRT1 in hUTC and PBMC.
  • MME CD10
  • ANPEP CD13
  • PTPRC CD45
  • DKK3 LAMB1, NRP1, GAPPD
  • HPRT1 HPRT1 in hUTC and PBMC.
  • GAPDH and HPRT all the genes tested show a greater abundance in hUTC as compared to human PBMC.
  • PTPRC CD45
  • a higher CT value is indicative of a lower amount of transcript.
  • PBMC is comprised of various types of blood cells
  • the gene expression profiles of select genes expressed in each of these cell types to that of hUTC were compared.
  • MME CD10
  • ANPEP CD13
  • T-cells and macrophages T-cells and macrophages
  • the cells were harvested in the exponential phase and subsequently, fixed and permeablized using a kit purchased from BD Bioscience. An aliquot of this preparation was then incubated with antibodies against selected markers identified to be present on hUTC surface, namely, CD10, CD13, CD45, NRP1, and LAMP1. After removing excess antibody, the cells were incubated with a fluorescently labeled secondary antibody. Cells were then analyzed by a flow cytometer.
  • Figures 4A to 4C The results for the flow cytometry assay for the detection of cell surface, plasma membrane, and intracellular markers are shown in Figures 4A to 4C .
  • Figure 4A shows the cell surface markers that were tested using hUTC with the top panel being the control.
  • Figure 4B shows the cell surface markers that were tested using PBMC with the top panel being the control.
  • Figure 4C shows the results for a flow cytometry assay for the detection of the internal markers DKK3 and LAMP1 in PBMC and hUTC.
  • the cell surface protein gene markers could be used to detect intact, live hUTC in mixed samples containing PBMCs in serum (see Figures 4A and 4B ). Even though NRP1 is transcribed at higher levels in hUTC as compared to that of human PBMC ( Figure 3 ), NRP1 could not be detected in hUTC by flow cytometry (see Figure 4A ). With respect to CD45, as expected, high levels of CD45 were being expressed on the surface of live human PBMC only (see Figure 4B ).
  • LAMP1 and DKK3 from the list of differentially expressed genes, Table 1-4 whose expression is higher in hUTC as compared to human PBMC were also examined (see Figure 4C ). These markers can be used as additional confirmation for hUTC identity.
  • Table 3-1 Difference between hUTC and PBMC with respect to percent positive cells as assayed by flow cytometry.
  • Table 3-2 summarizes the differences between hUTC and PBMC as assayed by RT-PCR in Example 2 above and by flow cytometry in the instant Example.
  • Nd means Not determined.
  • the two types of cells were mixed in 1 ml of human serum at room temperature. Immediately thereafter, aliquots of the mixture were analyzed by flow cytometry using CD10 as a marker for hUTC and CD45 as a marker for PBMC.
  • the concentration of hUTC ranged from 1,500 to 1,700 cells/ml and that of human PBMC was 1 million cells/ml.
  • the concentration of hUTC ranged from 1,700 to 110,000 cells/ml and that of human PBMC was 1 million cells/ml.
  • Umbilical cell isolation Umbilical cords were obtained from National Disease Research Interchange (NDRI, Philadelphia, PA). The tissues were obtained following normal deliveries. The cell isolation protocols were performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, CA) in the presence of penicillin at 100 U/ml, streptomycin at 100 mg/ml and amphotericin B at 0.25 ⁇ g/ml (Invitrogen Carlsbad, CA).
  • PBS phosphate buffered saline
  • the tissues were then mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 ml of medium (DMEM-low glucose or DMEM-high glucose; Invitrogen) until the tissue was minced into a fine pulp.
  • the chopped tissues were transferred to 50 ml conical tubes (approximately 5 g of tissue per tube).
  • the tissue was then digested in either DMEM-low glucose medium or DMEM-high glucose medium, each containing penicillin at 100 U/ml, streptomycin at 100 mg/ml, amphotericin B at 0.25 ⁇ g /ml and the digestion enzymes.
  • C:D collagenase and dispase
  • collagenase Sigma, St Louis, MO
  • dispase Invitrogen
  • C:D:H collagenase, 500 U/ml; dispase, 50 U/ml; and hyaluronidase (Sigma), 5 U/ml, in DMEM-Low glucose).
  • the conical tubes containing the tissue, medium and digestion enzymes were incubated at 37°C in an orbital shaker (Environ, Brooklyn, NY) at 225 rpm for 2 hours.
  • the tissues were centrifuged at 150 x g for 5 minutes, the supernatant was aspirated.
  • the pellet was resuspended in 20 ml of growth medium (DMEM:Low glucose (Invitrogen), 15% (v/v) fetal bovine serum (FBS; defined fetal bovine serum; Lot #AND18475; Hyclone, Logan, UT), 0.001% (v/v) 2-mercaptoethanol (Sigma), penicillin at 100 U/ml, streptomycin at 100 ⁇ g/ ml, and amphotericin B at 0.25 ⁇ g/ml (each from Invitrogen, Carlsbad, CA)).
  • DMEM Low glucose
  • FBS defined fetal bovine serum
  • Lot #AND18475 Hyclone, Logan, UT
  • 2-mercaptoethanol Sigma
  • penicillin 100 U/ml
  • streptomycin 100 ⁇ g/ ml
  • amphotericin B at 0.25 ⁇ g/ml (
  • the cell suspension was filtered through a 70 ⁇ m nylon BD FALCON Cell Strainer (BD Biosciences, San Jose, CA). An additional 5 ml rinse comprising growth medium was passed through the strainer. The cell suspension was then passed through a 40- ⁇ m nylon cell strainer (BD Biosciences, San Jose, CA) and chased with a rinse of an additional 5 ml of growth medium.
  • BD FALCON Cell Strainer BD Biosciences, San Jose, CA
  • the filtrate was resuspended in growth medium (total volume 50 ml) and centrifuged at 150 x g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 ml of fresh growth medium. This process was repeated twice more.
  • the cells isolated from umbilical cord tissues were seeded at 5,000 cells/cm 2 onto gelatin-coated T-75 flasks (Corning Inc., Corning, NY) in growth medium. After two days, spent medium and unadhered cells were aspirated from the flasks. Adherent cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with growth medium and allowed to grow to confluence (about 10 days from passage 0 to passage 1). On subsequent passages (from passage 1 to 2 etc.), cells reached sub-confluence (75-85% confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5,000 cells/cm 2 . Cells were grown in a humidified incubator with 5% carbon dioxide at 37°C.
  • cells were isolated from postpartum tissues in DMEM-low glucose medium after digestion with LIBERASE (2.5 mg/ml, Blendzyme 3; Roche Applied Sciences, Indianapolis, IN) and hyaluronidase (5 U/ml, Sigma). Digestion of the tissue and isolation of the cells was as described for other protease digestions above, however, the LIBERASE/hyaluronidase mixture was used instead of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.
  • Enzymes compared for digestion included: i) collagenase; ii) dispase; iii) hyaluronidase; iv) collagenase : dispase mixture (C:D); v) collagenase:hyaluronidase mixture (C:H); vi) dispase:hyaluronidase mixture (D:H); and vii) collagenase:dispase:hyaluronidase mixture (C:D:H). Differences in cell isolation utilizing these different enzyme digestion conditions were observed (see Table 5-1).
  • umbilical cord was sliced and washed with growth medium to dislodge the blood clots and gelatinous material.
  • the mixture of blood, gelatinous material and growth medium was collected and centrifuged at 150 x g. The pellet was resuspended and seeded onto gelatin coated flasks in growth medium. From these experiments, a cell population was isolated that readily expanded.
  • NDRI cord blood samples obtained from NDRI.
  • the isolation protocol used was that of International Patent Application PCT/US2002/029971 by Ho et al.
  • Samples (50 ml and 10.5 ml, respectively) of umbilical cord blood (NDRI, Philadelphia PA) were mixed with lysis buffer (filter-sterilized 155 mM ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 mM EDTA buffered to pH 7.2 (all components from Sigma, St. Louis, MO)).
  • lysis buffer filter-sterilized 155 mM ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 mM EDTA buffered to pH 7.2 (all components from Sigma, St. Louis, MO)
  • Cells were lysed at a ratio of 1:20 cord blood to lysis buffer.
  • the resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature.
  • the lysate was centrifuged (10 minutes at 200 x g).
  • the cell pellet was resuspended in Complete Minimal Essential Medium (Gibco, Carlsbad CA) containing 10% fetal bovine serum (Hyclone, Logan UT), 4 mM glutamine (Mediatech Herndon, VA), penicillin at 100 U/ml and streptomycin at 100 ⁇ g/ml (Gibco, Carlsbad, CA).
  • the resuspended cells were centrifuged (10 minutes at 200 x g), the supernatant was aspirated, and the cell pellet was washed in complete medium.
  • T75 flasks (Corning, NY), T75 laminin-coated flasks, or T175 fibronectin-coated flasks (both Becton Dickinson, Bedford, MA).
  • cells were digested in growth medium with or without 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO), using the enzyme combination of C:D:H, according to the procedures provided above. All cells were grown in the presence of penicillin at 100 U/ml and streptomycin at 100 ⁇ g/ml. Under all tested conditions, cells attached and expanded well between passage 0 and 1 (Table 4-2). Cells in conditions 5-8 and 13-16 were demonstrated to proliferate well up to 4 passages after seeding, at which point they were cryopreserved.
  • Table 5-2 Isolation and culture expansion of postpartum cells under varying conditions Condition Medium 15% FBS BME Gelatin 20% O 2 Growth Factors 1 DMEM-Lg Y Y Y Y N 2 DMEM-Lg Y Y Y N (5%) N 3 DMEM-Lg Y Y N Y N 4 DMEM-Lg Y Y N N (5%) N 5 DMEM-Lg N (2%) Y N (Laminin) Y EGF/FGF (20 ng/ml) 6 DMEM-Lg N (2%) Y N (Laminin) N (5%) EGF/FGF (20 ng/ml) 7 DMEM-Lg N (2%) Y N (Fibronectin) Y PDGF/VEGF 8 DMEM-Lg N (2%) Y N (Fibronectin) N (5%) PDGF/VEGF 9 DMEM-Lg Y N Y Y N 10 DMEM-Lg Y N Y N (5%) N 11 DMEM-
  • the preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
  • Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue, which adhere and grow under the conditions used, may be due to cells being released during the dissection process.
  • Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Human cells used in cell therapy should have a normal number (46) of chromosomes with normal structure. To identify umbilicus-derived cell lines that are homogeneous and free from cells of non-umbilical tissue origin, karyotypes of cell samples were analyzed.
  • UTC from postpartum tissue of a male neonate were cultured in growth media.
  • Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X,X).
  • Cells were seeded at 5,000 cells per square centimeter in growth medium in a T25 flask (Corning, Corning, NY) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with growth media. Samples were delivered to a clinical cytogenetics lab by courier (estimated lab to lab transport time is one hour). Chromosome analysis was performed by the Center for Human & Molecular Genetics at the New Jersey Medical School, Newark, NJ.
  • Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.
  • Chromosome analysis identified umbilicus-derived UTC whose karyotypes appear normal as interpreted by a clinical cytogenetic laboratory.
  • Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
  • Characterization of cell surface proteins or "markers" by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions. Postpartum cell lines isolated from the umbilicus were characterized by flow cytometry, providing a profile for the identification of these cell lines.
  • Cells were cultured in growth medium, in plasma-treated T75, T150, and T225 tissue culture flasks (Corning, Corning, NY) until confluent.
  • the growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin (Sigma, St. Louis, MO) for 20 minutes at room temperature.
  • Adherent cells in flasks were washed in phosphate buffered saline (PBS); (Gibco, Carlsbad, MO) and detached with trypsin/EDTA (Gibco). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1x10 7 1 ml.
  • antibody to the cell surface marker of interest was added to 100 ⁇ l of cell suspension and the mixture was incubated in the dark for 30 minutes at 4 °C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were resuspended in 500 ⁇ l PBS and analyzed by flow cytometry. Flow cytometry analysis was performed with a FACS calibur instrument (Becton Dickinson, San Jose, CA).
  • Umbilicus-derived cells were analyzed at passages 8, 15, and 20.
  • umbilical cord tissue-derived cells from different donors were compared to each other.
  • Umbilicus-derived cells cultured on gelatin-coated flasks were also compared to umbilicus-derived cells cultured on uncoated flasks.
  • Umbilical cord-derived cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control.
  • Umbilical cord-derived cells isolated from separate donors analyzed by flow cytometry each showed positive for the production of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for the production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control.
  • the umbilical cord-derived cells expanded on gelatin-coated and uncoated flasks analyzed by flow cytometry were all positive for the production of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha, and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for the production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
  • umbilical cord-derived cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C; and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ.
  • This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, digestion enzymes, and placental layer.
  • Oligonucleotide arrays were used to compare gene expression profiles of umbilicus-derived and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
  • Postpartum tissue-derived cells Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, PA) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 5 after digestion with a C:D:H mixture. The cells were cultured in growth medium on gelatin-coated plastic tissue culture flasks. The cultures were incubated at 37° C with 5% CO 2 .
  • Fibroblasts Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, MD; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F12 medium (Invitrogen, Carlsbad, CA) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen)). The cells were grown on standard tissue-culture treated plastic.
  • hMSC Human Mesenchymal Stem Cells
  • Cambrex Incorporated Walkersville, MD; Lot numbers 2F1655, 2F1656 and 2F1657) and cultured according to the manufacturer's specifications in MSCGM Media (Cambrex). The cells were grown on standard tissue cultured plastic at 37 °C with 5% CO 2 .
  • Human Iliac Crest Bone Marrow Cells (ICBM). Human iliac crest bone marrow was received from NDRI with patient consent. The marrow was processed according to the method outlined by Ho, et al. ( WO03/025149 ). The marrow was mixed with lysis buffer (155 mM NH 4 Cl, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 x g.
  • lysis buffer 155 mM NH 4 Cl, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2
  • the supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine.
  • the cells were centrifuged again and the cell pellet was resuspended in fresh medium.
  • the viable mononuclear cells were counted using trypan blue exclusion (Sigma, St. Louis, MO).
  • the mononuclear cells were seeded in plastic tissue culture flasks at 5 x 10 4 cells/cm 2 .
  • the cells were incubated at 37 °C with 5% CO 2 at either standard atmospheric O 2 or at 5% O 2 .
  • Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culturing. The adherent cells were maintained in culture.
  • the biotin-labeled cRNA was hybridized with Affymetrix GENECHIP HG-U133A oligonucleotide arrays (Affymetrix, Santa Clara, CA). The hybridizations and data collection were performed according to the manufacturer's specifications. Data analysis was performed using "Significance Analysis of Microarrays" (SAM) version 1.21 computer software ( Tusher, V.G. et al., 2001, Proc. Natl. Acad. Sci. USA 98: 5116-5121 ). Licenses for the analysis software are available through the Office of Technology Licensing, Stanford University, and more information is available on the World Wide Web at Professor Tibshirani's web site in the Dep't of Statistics, Stanford University.
  • SAM Signal Analysis of Microarrays
  • Table 8-1 Cells analyzed by the microarray study.
  • the data were evaluated by principle component analysis with SAM software as described above.
  • the analysis revealed 290 genes that were expressed in different relative amounts in the cells tested. This analysis provided relative comparisons between the populations.
  • Table 8-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs.
  • the Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types.
  • the Euclidean distance is inversely proportional to similarity between the expression of the 290 genes.
  • the Euclidean distance was calculated for the cell types using these 290 genes expressed differentially between the cell types. Similarity between the cells is inversely proportional to the Euclidean distance.
  • Table 8-2 The Euclidean Distances for the Cell Pairs.
  • Tables 8-3, 8-4, and 8-5 show the expression of genes increased in placenta-derived cells (Table 8-3), increased in umbilical cord-derived cells (Table 8-4), and reduced in umbilical cord and placenta-derived cells (Table 8-5)
  • Table 8-3 Genes which are specifically increased in expression in the placenta-derived cells as compared to the other cell lines assayed.
  • Tables 8-6, 8-7, and 8-8 show the expression of genes increased in human fibroblasts (Tab le 8-6), ICBM cells (Table 8-7), and MSCs (Table 8-8).
  • Table 8-6 Genes which were increased in expression in fibroblasts as compared to the other cell lines assayed.
  • interleukin 26 •maltase-glucoamylase ( ⁇ -glucosidase) •nuclear receptor subfamily 4, group A, member 2 •v-fos FBJ murine osteosarcoma viral oncogene homolog •hypothetical protein
  • DC42 •nuclear receptor subfamily 4, group A, member 2 •FBJ murine osteosarcoma viral oncogene homolog
  • B •WNT1 inducible signaling pathway protein 1 •MCF.2 cell line derived transforming sequence •potassium channel, subfamily K, member 15 •cartilage paired-class homeoprotein 1 •Homo sapiens cDNA FLJ12232 fis, clone MAMMA1001206 •Homo sapiens cDNA FLJ34668 fis, clone LIVER2000775 •jun B proto-oncogene •B-cell CLL/lymphoma 6 (zinc finger protein 51) •zinc finger protein 36, C3H type, homolog (mouse
  • This example was performed to provide a molecular characterization of the cells derived from umbilical cord and placenta.
  • This analysis included cells derived from three different umbilical cords and three different placentas.
  • the study also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of iliac crest bone marrow cells.
  • the mRNA that was expressed by these cells was analyzed on a GENECHIP oligonucleotide array that contained oligonucleotide probes for 22,000 genes.
  • transcripts for 290 genes were present in different amounts in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta-derived and umbilical cord tissue-derived cells.
  • the phenotypes of cells found within human umbilical cord tissue were analyzed by immunohistochemistry.
  • anti-human GROalpha-PE (1:100; Becton Dickinson, Franklin Lakes, NJ)
  • anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA)
  • anti-human oxidized LDL receptor 1 ox-LDL R1; 1:100; Santa Cruz Biotech
  • anti-human NOGO-A (1:100; Santa Cruz Biotech).
  • Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, CA) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 ⁇ m thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
  • Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord (data not shown). In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord. CD34 positive (CD34 + ) cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery and vein, but not contained within the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
  • Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD 34 are expressed in cells within human umbilical cord. Based on in vitro characterization studies showing that only vimentin and alpha-smooth muscle actin are expressed, the data suggests that the current process of umbilical cord-derived cell isolation harvests a subpopulation of cells or that the cells isolated change expression of markers to express vimentin and alpha-smooth muscle actin.
  • HGF hepatocyte growth factor
  • MCP-1 monocyte chemotactic protein 1
  • IL-8 interleukin-8
  • KGF keratinocyte growth factor
  • bFGF basic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • tissue inhibitor of matrix metalloproteinase 1 TPO
  • tissue inhibitor of matrix metalloproteinase 1 TPO
  • ANG2 angiopoietin 2
  • PDGFbb platelet derived growth factor
  • TPO thrombopoietin
  • HB-EGF stromal-derived factor 1alpha
  • SDF-1alpha neurotrophic/neuroprotective activity
  • BDNF brain-derived neurotrophic factor
  • IL-6 interleukin-6
  • GCP-2 granulocyte chemotactic protein-2
  • TGFbeta2 transforming growth factor beta2
  • chemokine activity microphage inflammatory protein 1alpha
  • MIP1beta macrophage inflammatory protein 1 beta
  • MCP-1 monocyte chemoattractant-1
  • Rantes regulated on activation, normal T cell expressed and secreted
  • 1309 thymus and activation-regulated chemokine (TARC); Eotaxin; macrophage-derived chemokine (MDC); and (IL-8).
  • the medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 U/ml) and streptomycin (50 ⁇ g/ml, Gibco)) for 8 hours.
  • DMEM-low glucose (Gibco) 0.1% (w/v) bovine serum albumin (Sigma), penicillin (50 U/ml) and streptomycin (50 ⁇ g/ml, Gibco)
  • Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000 x g for 5 minutes and stored at -20°C.
  • the cells were washed with phosphate-buffered saline (PBS) and detached using 2 ml trypsin/EDTA (Gibco). Trypsin activity was inhibited by addition of 8 ml growth medium. The cells were centrifuged at 150 x g for 5 minutes. The supernatant was removed, and the cells were resuspended in 1 ml Growth Medium. The cell number was estimated with a hemocytometer.
  • PBS phosphate-buffered saline
  • EDTA Gibco
  • Trypsin activity was inhibited by addition of 8 ml growth medium.
  • the cells were centrifuged at 150 x g for 5 minutes. The supernatant was removed, and the cells were resuspended in 1 ml Growth Medium. The cell number was estimated with a hemocytometer.
  • Chemokines (MIP1alpha, MIP1beta, MCP-1, Rantes, 1309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMP1, ANG2, PDGFbb, TPO, HB-EGF were measured using SearchLight Proteome Arrays (Pierce Biotechnology Inc.).
  • the Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to sixteen proteins per well. The arrays are produced by spotting a 2 x 2, 3 x 3, or 4 x 4 pattern of four to sixteen different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture the chemiluminescent signal generated at each spot within each well of the plate. The signal generated at each spot is proportional to the amount of target protein in the original standard or sample.
  • MCP-1 and IL-6 were secreted by umbilicus-derived PPDCs and dermal fibroblasts (Table 10-1). SDF-1alpha and GCP-2 were secreted by fibroblasts. GCP-2 and IL-8 were secreted by umbilicus-derived PPDCs. TGF-beta2 was not detected from either cell type by ELISA. Table 10-1.
  • TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1beta, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were secreted from umbilicus-derived PPDCs (Tables 10-2 and 10-3). No Ang2, VEGF, or PDGFbb were detected. Table 10-2.
  • Umbilicus-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration or protection.
  • Telomerase functions to synthesize telomere repeats that serve to protect the integrity of chromosomes and to prolong the replicative life span of cells ( Liu, K, et al., PNAS, 1999; 96:5147-5152 ). Telomerase consists of two components, telomerase RNA template (hTER) and telomerase reverse transcriptase (hTERT). Regulation of telomerase is determined by transcription of hTERT but not hTER. Real-time polymerase chain reaction (PCR) for hTERT mRNA thus is an accepted method for determining telomerase activity of cells.
  • PCR Real-time polymerase chain reaction
  • telomerase production of human umbilical cord tissue-derived cells Human umbilical cord tissue-derived cells were prepared in accordance with the above Examples and the examples set forth in U.S. Patent No. 7,510,873 . Generally, umbilical cords obtained from National Disease Research Interchange (Philadelphia, Pa.) following a normal delivery were washed to remove blood and debris and mechanically dissociated. The tissue was then incubated with digestion enzymes including collagenase, dispase, and hyaluronidase in culture medium at 37°C. Human umbilical cord tissue-derived cells were cultured according to the methods set forth in the examples of the '012 application.
  • NTERA-2 cl.Dl pluripotent human testicular embryonal carcinoma (teratoma) cell line nTera-2 cells (NTERA-2 cl.Dl) ( See, Plaia et al., Stem Cells, 2006; 24(3):531-546 ) was purchased from ATCC (Manassas, Va.) and was cultured according to the methods set forth in U.S. Patent No. 7,510,873 .
  • PCR was performed on cDNA samples using the Applied Biosystems Assays-On-DemandTM (also known as TaqMan® Gene Expression Assays) according to the manufacturer's specifications (Applied Biosystems).
  • This commercial kit is widely used to assay for telomerase in human cells. Briefly, hTert (human telomerase gene) (Hs00162669) and human GAPDH (an internal control) were mixed with cDNA and TaqMan® Universal PCR master mix using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems). Thermal cycle conditions were initially 50°C for 2 minutes and 95°C for 10 minutes followed by 40 cycles of 95°C for 15 seconds and 60°C for 1 minute. PCR data was analyzed according to the manufacturer's specifications.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Wood Science & Technology (AREA)
  • Virology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Claims (13)

  1. Verfahren zum Nachweis zirkulierender allogener therapeutischer Zellen in einer Blutprobe, umfassend:
    a. Testen zirkulierender allogener therapeutischer Zellen und menschlicher mononukleärer Zellen aus peripherem Blut, um einen oder mehrere für allogene therapeutische Zellen positive(n) Marker und einen oder mehrere für mononukleäre Zellen aus peripherem Blut des Patienten positive(n) Marker zu identifizieren;
    b. Vergleichen des einen oder der mehreren für zirkulierende allogene therapeutische Zellen positiven Marker (s) und des einen oder der mehreren für mononukleäre Zellen aus peripherem Blut des Patienten positiven Marker(s), um einen oder mehrere einzigartige Marker zu identifizieren, der die allogenen therapeutischen Zellen von den mononukleären Zellen aus peripherem Blut des Patienten unterscheidet;
    c. Analysieren einer Blutprobe von einem Patienten, der mit allogenen therapeutischen Zellen behandelt wurde, unter Verwendung eines Assayverfahrens zum Nachweis des einen oder der mehreren für die zirkulierenden allogenen therapeutischen Zellen positiven Marker(s); und
    d. Unterscheiden zwischen den mononukleären Zellen aus peripherem Blut des Patienten und zirkulierenden allogenen therapeutischen Zellen auf der Basis des Nachweises des einen oder der mehreren einzigartigen Marker(s),
    wobei die zirkulierenden allogenen therapeutischen Zellen aus menschlichem Nabelschnurgewebe stammende Zellen sind und wobei der eine oder die mehreren für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen einzigartige(n) Marker CD10, CD13, NRP1, LAMP1, DKK3 oder LAMB1 umfasst/umfassen.
  2. Verfahren nach Anspruch 1, wobei der eine oder die mehreren für mononukleäre Zellen aus peripherem Blut des Patienten positive(n) Marker CD45 umfasst/umfassen.
  3. Verfahren nach Anspruch 1, wobei das Assayverfahren in Schritt b. aus der Gruppe ausgewählt ist, bestehend aus Durchflusszytometrie, ELISA, Immunhistochemie, Nukleinsäurenachweis, PCR und Kombinationen davon.
  4. Verfahren nach Anspruch 1, das außerdem das Durchführen eines Anreicherungsschritts zwischen den Schritten (c) und (d) umfasst.
  5. Verfahren nach Anspruch 4, wobei es sich bei dem Anreicherungsschritt um Magneteinfangtechnologie handelt.
  6. Verfahren nach Anspruch 1, wobei der Unterscheidungsschritt das Unterscheiden zwischen zirkulierenden allogenen therapeutischen Zellen, die dem Patienten verabreicht worden sind, und mononukleären Zellen aus peripherem Blut des Patienten umfasst.
  7. Verfahren nach Anspruch 1, wobei der Patient ein Mensch, ein nicht-menschlicher Primat, eine Maus, eine Ratte, ein Hamster, ein Meerschweinchen, ein Hund oder ein Schwein ist.
  8. Verfahren nach einem der Ansprüche 1-7, wobei der für mononukleäre Zellen aus peripherem Blut des Patienten positive Marker CD45 ist und der für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen positive Marker CD10 ist.
  9. Verfahren nach Anspruch 8, das weiterhin CD13 als den positiven Marker für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen umfasst.
  10. Verfahren nach Anspruch 8, das weiterhin eines oder mehrere aus NRP1, DKK3 und LAMP1 als den positiven Marker für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen umfasst.
  11. Verfahren zum Nachweis zirkulierender aus menschlichem Nabelschnurgewebe stammender Zellen in einer Blutprobe, das die folgenden Schritte umfasst:
    a. Isolieren der Fraktion der aus menschlichem Nabelschnurgewebe stammenden Zellen/mononukleären Zellen aus peripherem Blut aus einer Blutprobe, die zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen enthält;
    b. Entfernen jeglichen Plasmas;
    c. Analysieren der Fraktion der aus menschlichem Nabelschnurgewebe stammenden Zellen/mononukleären Zellen aus peripherem Blut mittels Durchflusszytometrie auf CD45 als einen positiven Marker für mononukleäre Zellen aus peripherem Blut und CD13 als einen positiven Marker für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen; und
    d. Nachweisen des Vorhandenseins der menschlichen mononukleären Zellen aus peripherem Blut und der zirkulierenden aus menschlichem Nabelschnurgewebe stammenden Zellen auf der Basis des Nachweises von CD45 als einem für menschliche mononukleäre Zellen aus peripherem Bluts positiven Marker und von CD13 als einem für aus menschlichem Nabelschnurgewebe stammende Zellen positiven Marker.
  12. Verfahren nach Anspruch 11, das weiterhin das Analysieren von CD10 als einen für zirkulierende aus menschlichem Nabelschnurgewebe stammende Zellen positiven Marker umfasst.
  13. Verfahren nach Anspruch 8 oder 11, wobei die zirkulierenden aus menschlichem Nabelschnurgewebe stammenden Zellen aus menschlichem Nabelschnurgewebe im Wesentlichen frei von Blut isoliert worden sind, zur Selbsterneuerung und zur Vermehrung in Kultur in der Lage sind, das Potenzial zur Differenzierung besitzen und die folgenden Merkmale aufweisen: (1) CD10, CD13, CD44, CD90 und HLA-ABC exprimieren; (2) CD31, CD34, CD45, HLA-DR und CD117 nicht exprimieren; (3) hTERT oder Telomerase nicht exprimieren; (4) den oxidiertes-Low-Density-Lipoprotein-Rezeptor 1, Reticulon, Chemokinrezeptorligand 3 und/oder Granulozytenchemotaktisches Protein exprimieren; und (4) im Vergleich zu einem menschlichen Fibroblasten, einer mesenchymalen Stammzelle oder einer Beckenkammknochenmarkzelle erhöhte Spiegel an Interleukin 8 oder Reticulon 1 exprimieren.
EP12815943.1A 2011-12-23 2012-12-20 Nachweis von zellen aus menschlichem nabelschnurgewebe Not-in-force EP2794854B1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PL12815943T PL2794854T3 (pl) 2011-12-23 2012-12-20 Wykrywanie ludzkich komórek pochodzących z tkanki pępowinowej

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161579710P 2011-12-23 2011-12-23
PCT/US2012/071072 WO2013096686A1 (en) 2011-12-23 2012-12-20 Detection of human umbilical cord tissue-derived cells

Publications (2)

Publication Number Publication Date
EP2794854A1 EP2794854A1 (de) 2014-10-29
EP2794854B1 true EP2794854B1 (de) 2018-06-20

Family

ID=47559711

Family Applications (1)

Application Number Title Priority Date Filing Date
EP12815943.1A Not-in-force EP2794854B1 (de) 2011-12-23 2012-12-20 Nachweis von zellen aus menschlichem nabelschnurgewebe

Country Status (17)

Country Link
US (2) US9611513B2 (de)
EP (1) EP2794854B1 (de)
JP (1) JP6301263B2 (de)
KR (1) KR101981450B1 (de)
CN (1) CN104837987B (de)
AU (1) AU2012358810B2 (de)
BR (1) BR112014015424A2 (de)
CA (1) CA2869681A1 (de)
ES (1) ES2676556T3 (de)
HK (2) HK1203550A1 (de)
MX (1) MX362198B (de)
PH (1) PH12014501445A1 (de)
PL (1) PL2794854T3 (de)
RU (1) RU2636220C2 (de)
SG (1) SG11201403465PA (de)
WO (1) WO2013096686A1 (de)
ZA (1) ZA201405407B (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2012358810B2 (en) 2011-12-23 2018-03-15 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue-derived cells
CN104678109A (zh) * 2015-03-06 2015-06-03 复旦大学附属中山医院 一种人源性的cd13可溶性蛋白的免疫组化试剂盒及其应用
CN105238751B (zh) * 2015-11-30 2021-07-23 深圳市合一康生物科技股份有限公司 一种脐带组织间充质干细胞的分离培养方法
CN109104869B (zh) 2016-04-27 2022-07-01 日本乐敦制药株式会社 表达细胞表面标记物的间充质干细胞、包含该间充质干细胞的药物组合物及其制备方法
WO2019036577A1 (en) * 2017-08-18 2019-02-21 University Of South Carolina DETECTION OF A MODIFICATION DERIVED FROM FUMARATE ESTER IN A TEST SAMPLE

Family Cites Families (215)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4018886A (en) 1975-07-01 1977-04-19 General Electric Company Diagnostic method and device employing protein-coated magnetic particles
US3970518A (en) 1975-07-01 1976-07-20 General Electric Company Magnetic separation of biological particles
US4267234A (en) 1978-03-17 1981-05-12 California Institute Of Technology Polyglutaraldehyde synthesis and protein bonding substrates
US4230685A (en) 1979-02-28 1980-10-28 Northwestern University Method of magnetic separation of cells and the like, and microspheres for use therein
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4452773A (en) 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
US4659678A (en) 1982-09-29 1987-04-21 Serono Diagnostics Limited Immunoassay of antigens
US4554088A (en) 1983-05-12 1985-11-19 Advanced Magnetics Inc. Magnetic particles for use in separations
US4795698A (en) 1985-10-04 1989-01-03 Immunicon Corporation Magnetic-polymer particles
US5597531A (en) 1985-10-04 1997-01-28 Immunivest Corporation Resuspendable coated magnetic particles and stable magnetic particle suspensions
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5863531A (en) 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5266480A (en) 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
CA1322262C (en) 1987-06-26 1993-09-21 Yoshito Ikada Artificial skin
NZ226750A (en) 1987-10-29 1990-09-26 Amrad Corp Ltd Immortalisation of neural precursor cells by introducing a retrovirus vector containing a myc-oncogene
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US20030032178A1 (en) 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
FR2646438B1 (fr) 1989-03-20 2007-11-02 Pasteur Institut Procede de remplacement specifique d'une copie d'un gene present dans le genome receveur par l'integration d'un gene different de celui ou se fait l'integration
US5437994A (en) 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5698271A (en) 1989-08-22 1997-12-16 Immunivest Corporation Methods for the manufacture of magnetically responsive particles
US5840580A (en) 1990-05-01 1998-11-24 Becton Dickinson And Company Phenotypic characterization of the hematopoietic stem cell
AU664976B2 (en) 1990-08-29 1995-12-14 Gene Pharming Europe Bv Homologous recombination in mammalian cells
US5342761A (en) 1990-10-01 1994-08-30 Research Development Foundation Oncofetal gene, gene product and uses therefor
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5286632A (en) 1991-01-09 1994-02-15 Jones Douglas H Method for in vivo recombination and mutagenesis
US6399369B1 (en) 1991-07-08 2002-06-04 Neurospheres Holdings Ltd. Multipotent neural stem cell cDNA libraries
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
WO1994000484A1 (en) 1992-06-22 1994-01-06 Young Henry E Scar inhibitory factor and use thereof
US5629147A (en) 1992-07-17 1997-05-13 Aprogenex, Inc. Enriching and identifying fetal cells in maternal blood for in situ hybridization
US5320962A (en) 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US20040224409A1 (en) 1992-09-25 2004-11-11 Laurent Pradier Recombinant adenoviruses coding for brain-derived neurotrophic factor (BDNF)
WO1994010331A1 (en) 1992-10-29 1994-05-11 The Australian National University Angiogenesis inhibitory antibodies
US5670483A (en) 1992-12-28 1997-09-23 Massachusetts Insititute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5955343A (en) 1992-12-28 1999-09-21 Massachusetts Institute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
WO1994025584A1 (en) 1993-04-28 1994-11-10 Johns Hopkins University School Of Medicine Chronic endothelial cell culture under flow
IL110589A0 (en) 1993-08-10 1994-11-11 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF- beta family
US6686198B1 (en) 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
DE4406073A1 (de) 1994-02-24 1995-08-31 Univ Ludwigs Albert Verfahren zur Herstellung von humanen, klonogenen Fibroblasten, Verfahren zur Gentransfizierung von Fibroblasten und so erhaltene Fibroblasten
US6703017B1 (en) 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
IL114397A0 (en) 1994-07-01 1995-10-31 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF-beta-family
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US5725493A (en) 1994-12-12 1998-03-10 Avery; Robert Logan Intravitreal medicine delivery
US5684032A (en) 1994-12-13 1997-11-04 Smithkline Beecham Corporation Compounds
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5906934A (en) 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5869079A (en) 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5641750A (en) 1995-11-29 1997-06-24 Amgen Inc. Methods for treating photoreceptors using glial cell line-derived neurotrophic factor (GDNF) protein product
US6200606B1 (en) 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
US5842477A (en) 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
CA2248549A1 (en) 1996-03-15 1997-09-18 Munin Corporation Extracellular matrix signalling molecules
US6497875B1 (en) 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US6358737B1 (en) 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6787355B1 (en) 1996-08-26 2004-09-07 Mcgill University Multipotent neural stem cells from peripheral tissues and uses thereof
US5731156A (en) * 1996-10-21 1998-03-24 Applied Imaging, Inc. Use of anti-embryonic hemoglobin antibodies to identify fetal cells
US5919702A (en) 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
EP2110431A1 (de) 1997-05-13 2009-10-21 Osiris Therapeutics, Inc. Knorpelregeneration unter Verwendung menschlicher mesenchymaler Stammzellen
AU8401498A (en) 1997-07-14 1999-02-10 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
US5902598A (en) 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
AU1615599A (en) 1997-12-02 1999-06-16 Zen Bio, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6059968A (en) 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
US6291240B1 (en) 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
IL137802A0 (en) 1998-02-12 2001-10-31 Immunivest Corp A method and kit for detecting rare cells in a mixed cell population
ES2237089T3 (es) 1998-03-13 2005-07-16 Osiris Therapeutics, Inc. Usos para celulas madre mesenquimatosas humanas no autologas.
US6171610B1 (en) 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
JP2002513545A (ja) 1998-05-07 2002-05-14 ザ ユニヴァーシティー オブ サウス フロリダ 脳および脊髄修復のためのニューロン源としての骨髄細胞
WO1999061587A1 (en) 1998-05-29 1999-12-02 Osiris Therapeutics, Inc. Human cd45+ and/or fibroblast + mesenchymal stem cells
US6323188B1 (en) 1998-07-01 2001-11-27 Donald L. Weissman Treatment and prevention of cardiovascular diseases, heart attack, and stroke, primary and subsequent, with help of aspirin and certain vitamins
US20040037818A1 (en) 1998-07-30 2004-02-26 Brand Stephen J. Treatment for diabetes
DE69941510D1 (de) 1998-08-10 2009-11-19 Us Gov Nat Inst Health Differenzierung von nicht-insulin in insulin-produzierende zellen durch glp-1 und exendin-4 und dessen verwendung
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
US6284245B1 (en) 1998-08-25 2001-09-04 Diacrin, Inc. Neural retinal cells and retinal pigment epithelium cells and their use in treatment of retinal disorders
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
MXPA01007810A (es) 1999-02-04 2003-06-04 Univ Mcgill Plataforma para la diferenciacion de celulas.
US6326201B1 (en) 1999-02-10 2001-12-04 Curis, Inc. Pancreatic progenitor cells, methods and uses related thereto
ES2234523T3 (es) * 1999-03-01 2005-07-01 Pfizer Products Inc. Acidos oxamicos que contienen ciano y sus derivados como ligandos de receptores tiroideos.
US20030007954A1 (en) 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
CA2370330C (en) 1999-04-16 2009-01-20 Wm. Marsh Rice University Biodegradable poly(propylene fumarate) networks cross linked with poly(propylene fumarate)-diacrylate macromers
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
US6372494B1 (en) 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
WO2000073421A2 (en) 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6355699B1 (en) 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US7621606B2 (en) 2001-08-27 2009-11-24 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US6333029B1 (en) 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
WO2001011011A2 (en) 1999-08-05 2001-02-15 Mcl Llc Multipotent adult stem cells and methods for isolation
US6555374B1 (en) 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
JP2003509374A (ja) 1999-09-14 2003-03-11 チルドレンズ メディカル センター コーポレーション 骨髄細胞を用いる筋ジストロフィーの治療方法
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
EP1099754A1 (de) 1999-11-10 2001-05-16 Universiteit Leiden Mesenchym-Stammzellen und/oder Vorlaüferzellen, ihre Isolierung und ihre Verwendung
US20030082155A1 (en) 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20020164307A1 (en) 1999-12-06 2002-11-07 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
AU778929B2 (en) 1999-12-06 2004-12-23 General Hospital Corporation, The Pancreatic stem cells and their use in transplantation
WO2001053503A1 (en) 2000-01-18 2001-07-26 Cornell Research Foundation, Inc. Neural progenitor cells from hippocampal tissue and a method for isolating and purifying them
US7544509B2 (en) 2000-01-24 2009-06-09 Mcgill University Method for preparing stem cell preparations
US6610535B1 (en) 2000-02-10 2003-08-26 Es Cell International Pte Ltd. Progenitor cells and methods and uses related thereto
ATE473751T1 (de) 2000-02-11 2010-07-15 Schepens Eye Res Inst Isolierung und transplantation von retinalen stammzellen
AU2001236854B2 (en) 2000-02-11 2005-04-14 Philadelphia Health And Education Corporation Differentiation of bone marrow cells into neuronal cells and uses therefor
US7160724B2 (en) 2000-03-09 2007-01-09 University Of South Florida Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US6673606B1 (en) 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
AU2001264598B2 (en) 2000-05-12 2006-07-06 University Of Utah Research Foundation Compositions and methods for tissue dedifferentiation and regeneration
US8273570B2 (en) 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US6759039B2 (en) 2000-06-30 2004-07-06 Amcyte, Inc. Culturing pancreatic stem cells having a specified, intermediate stage of development
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
WO2002026941A2 (en) 2000-09-29 2002-04-04 Kooy Derek V D Primitive neural stem cells and method for differentiation of stem cells to neural cells
AU2002230669A1 (en) 2000-11-06 2002-05-15 The Salk Institute For Biological Studies Postmortem stem cells
IL156234A0 (en) 2000-11-30 2004-01-04 Stemron Inc Isolated homozygous stem cells, differentiated cells derived therefrom, and materials and methods for making and using same
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
EP3299452A1 (de) 2000-12-06 2018-03-28 Anthrogenesis Corporation Verfahren zum sammeln plazentärer stammzellen
US6599323B2 (en) 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
JP2005503759A (ja) 2001-01-24 2005-02-10 アメリカ合衆国 幹細胞の膵臓内分泌細胞への分化方法
DE60230593D1 (de) 2001-02-06 2009-02-12 Massachusetts Inst Technology Peptidgerüstverkapselung von gewebszellen und verwendungen davon
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
IL157332A0 (en) 2001-02-14 2004-02-19 Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
EP2338983B1 (de) 2001-02-14 2015-10-07 Anthrogenesis Corporation Regeneration und Repopulation von dezellularisierten Geweben und Kadaverorganen durch Stammzellen.
CA2438153C (en) 2001-02-14 2015-06-02 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
CA2442177A1 (en) 2001-03-29 2002-10-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US20050054102A1 (en) 2001-04-19 2005-03-10 Anna Wobus Method for differentiating stem cells into insulin-producing cells
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
US20030022369A1 (en) 2001-05-18 2003-01-30 Helen Fillmore Differentiation of specialized dermal and epidermal cells into neuronal cells
JP2005515753A (ja) 2001-05-25 2005-06-02 サイセラ,インコーポレイテッド 幹細胞分化
US7632680B2 (en) 2001-08-08 2009-12-15 Levesque Biosciences, Inc. Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
US20030211603A1 (en) 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
US7863012B2 (en) 2004-02-17 2011-01-04 Veridex, Llc Analysis of circulating tumor cells, fragments, and debris
US20030104997A1 (en) 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
US20050064587A1 (en) 2001-09-07 2005-03-24 Lawrence Rosenberg Pancreatic small cells and uses thereof
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
EP1298201A1 (de) 2001-09-27 2003-04-02 Cardion AG Verfahren zur Herstellung von Zellen, die einen pankreatischen Beta Inselzellen gleichen Status aufweisen
EP1446477A4 (de) 2001-09-28 2006-06-07 Es Cell Int Pte Ltd Verfahren der derivatisierung und vermehrung undifferenzierter humaner embryonaler stammzellen (hes-zellen) auf feeder-freien substraten und menschlichen feeder-schichten
WO2003033697A1 (en) 2001-10-18 2003-04-24 Ixion Biotechnology, Inc. Conversion of liver stem and progenitor cells to pancreatic functional cells
US7129034B2 (en) 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
AU2002359390A1 (en) 2001-11-09 2003-05-19 Artecel Sciences, Inc. Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
ATE438708T1 (de) 2001-11-15 2009-08-15 Childrens Medical Center Verfahren zur isolierung, expansion und differenzierung fötaler stammzellen aus chorionzotte, fruchtwasser und plazenta und therapeutische verwendungen davon
JP3728750B2 (ja) 2001-11-22 2005-12-21 ニプロ株式会社 培養皮膚及びその製造方法
ES2377099T3 (es) 2001-12-04 2012-03-22 Organogenesis Inc. Células cultivadas procedentes de islotes pancreáticos
US20030109036A1 (en) 2001-12-06 2003-06-12 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
WO2003050249A2 (en) 2001-12-07 2003-06-19 Geron Corporation Islet cells from human embryonic stem cells
JP3934539B2 (ja) 2001-12-12 2007-06-20 独立行政法人科学技術振興機構 胎盤等由来の成体又は生後組織の前駆細胞
US20030113910A1 (en) 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
ATE464373T1 (de) 2001-12-21 2010-04-15 Mount Sinai Hospital Corp Zelluläre zusammensetzungen und verfahren zur deren bereitung und verwendung
US7101546B2 (en) 2001-12-21 2006-09-05 Amcyte, Inc. In situ maturation of cultured pancreatic stem cells having a specified, intermediate stage of development
JP2005512593A (ja) 2001-12-28 2005-05-12 セルアーティス アーベー 多能性のヒト胚盤胞由来幹細胞株の樹立方法
EP1471970A4 (de) 2002-01-14 2006-08-02 Univ Illinois Neue multipotente säugetier-stammzellen und zusammensetzungen, verfahren zur herstellung und verfahren zu ihrer verabreichung
US20030162290A1 (en) 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
AU2003217357A1 (en) 2002-02-07 2003-09-02 The Research Foundation Of The State University Of New York Generation of new insulin cells from progenitor cells present in adult pancreatic islets
KR101176146B1 (ko) 2002-02-13 2012-08-22 안트로제네시스 코포레이션 산후 포유류 태반으로부터 유래한 배아-유사 줄기 세포와그 세포를 사용한 용도 및 치료방법
WO2003070189A2 (en) 2002-02-15 2003-08-28 Cornell Research Foundation, Inc. Enhancing neurotrophin-induced neurogenesis by endogenous neural progenitor cells by concurrent overexpression of brain derived neurotrophic factor and an inhibitor of a pro-gliogenic bone morphogenetic protein
CN1281739C (zh) 2002-02-19 2006-10-25 美迪宝斯特有限公司 从脐带血中分离并扩大培养间充质干细胞/祖细胞及将脐带血来源的间充质干细胞/祖细胞分化成各种间充质组织的方法
US7029915B2 (en) 2002-02-22 2006-04-18 University Of Florida Research Foundation, Inc. Method for differentiating rat hepatic stem cells to insulin-producing cells
US7736892B2 (en) 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20030161818A1 (en) 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7150990B2 (en) 2002-03-06 2006-12-19 Reprocell, Inc. Self-renewing pluripotent hepatic stem cells
JPWO2003080822A1 (ja) 2002-03-27 2005-07-28 ニプロ株式会社 胎盤由来の間葉系細胞およびその医学的用途
EP1538913A2 (de) 2002-04-12 2005-06-15 Celgene Corporation Modulation von stamm- und vorläuferzelldifferenzierung, analysen und verwendungen davon
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
JP4136434B2 (ja) 2002-04-17 2008-08-20 進 清野 インスリン産生細胞の誘導
JP2005523328A (ja) 2002-04-19 2005-08-04 ユニバーシティ オブ ピッツバーグ オブ ザ コモンウェルス システム オブ ハイヤー エデュケーション 胎盤由来の幹細胞及びその使用
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
JP2005526838A (ja) 2002-04-25 2005-09-08 ウイスコンシン アラムナイ リサーチ フオンデーシヨン パーキンソン病および他の神経変性疾患を治療するためのgdnf分泌ヒト神経幹細胞の使用
US20040029269A1 (en) 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
AU2003233119A1 (en) 2002-05-08 2003-11-11 Neuronova Ab Modulation of neural stem cells with s1p or lpa receptor agonists
EP1507848A4 (de) 2002-05-28 2005-11-23 Becton Dickinson Co Expansion und transdifferenzierung menschlicher azinuszellen
MXPA04011851A (es) 2002-05-30 2005-03-31 Celgene Corp Metodos para utilizar inhibidores de jnk o mkk para modular diferenciacion de celula y para tratar desordenes mieloproliferativos y sindromes mielodispl??sticos.
EP1511838A4 (de) 2002-06-07 2007-01-10 Es Cell Int Pte Ltd Verfahren zur steuerung der differenzierung in stammzellen
WO2003104423A2 (en) 2002-06-11 2003-12-18 Roy Ogle Meningeal-derived stem cells
US7285415B2 (en) 2002-07-11 2007-10-23 The Regents Of The University Of California Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US20050249731A1 (en) 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and formation
US7390659B2 (en) 2002-07-16 2008-06-24 The Trustees Of Columbia University In The City Of New York Methods for inducing differentiation of embryonic stem cells and uses thereof
US6621074B1 (en) 2002-07-18 2003-09-16 Perseptive Biosystems, Inc. Tandem time-of-flight mass spectrometer with improved performance for determining molecular structure
US7527968B2 (en) 2002-07-29 2009-05-05 Robarts Research Institute Regeneration initiating cells
AU2003257938A1 (en) 2002-07-29 2004-02-16 Es Cell International Pte Ltd. Multi-step method for differentiation of insulin positive, glucose
WO2004016747A2 (en) 2002-08-14 2004-02-26 University Of Florida Bone marrow cell differentiation
AU2003265856A1 (en) 2002-08-28 2004-03-19 University Of Florida Neurogenesis from hepatic stem cells
AU2003268534A1 (en) 2002-09-06 2004-03-29 Amcyte Inc. Cd56 positive human adult pancreatic endocrine progenitor cells
US9969977B2 (en) 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
JP4571387B2 (ja) 2003-02-07 2010-10-27 宣男 櫻川 ヒト羊膜由来サイドポピュレーション細胞及びその用途
ATE478941T1 (de) 2003-02-11 2010-09-15 John E Davies Vorläuferzellen aus der wharton sulze von humanen nabelschnüren
US20050009114A1 (en) * 2003-05-15 2005-01-13 Board Of Regents, The University Of Texas System Identification and quantification of organ-specific proteins derived from human allogeneic cells using proteomics
WO2004111207A1 (en) 2003-06-09 2004-12-23 Centelion Multicellular in vitro model of angiogenesis and methods for using the same
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
ES2600555T3 (es) 2003-06-27 2017-02-09 DePuy Synthes Products, Inc. Reparación y regeneración de tejido ocular usando células derivadas del post parto
US20060223177A1 (en) 2003-06-27 2006-10-05 Ethicon Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
CN101416059A (zh) 2003-07-17 2009-04-22 格洛伯塞尔解决方案公司 自动化细胞培养***及方法
ITRM20030386A1 (it) * 2003-08-05 2005-02-06 Istituto Naz Per Le Malattie Infettive Lazz Metodo e test diagnostici basati sull'analisi citofluorimetrica dei linfociti t antigene-specifici.
WO2005021738A1 (en) 2003-08-29 2005-03-10 Regents Of The University Of Minnesota Kidney derived stem cells and methods for their isolation, differentiation and use
US20050089513A1 (en) 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
WO2006083394A2 (en) 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
EP1838842A2 (de) 2004-12-23 2007-10-03 Ethicon, Incorporated Behandlung osteochondraler krankheiten unter verwendung postpartum gewonnener zellen und von produkten davon
WO2006071777A2 (en) 2004-12-23 2006-07-06 Ethicon Incorporated Soft tissue repair and regeneration using postpartum-derived cells and cell products
CA2602684C (en) 2005-03-31 2014-07-22 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US20110052545A1 (en) 2005-05-05 2011-03-03 Katalin Vehmeyer Regeneration system, its production and use
WO2006138275A2 (en) * 2005-06-13 2006-12-28 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
EP1943335A2 (de) 2005-11-02 2008-07-16 Georg-August-Universität Göttingen Zusammensetzungen und verfahren zur herstellung pluripotenter zellen aus adultem testis
DK2471903T3 (en) 2005-12-29 2018-05-28 Anthrogenesis Corp Placenta stem cell populations
CA2572530A1 (en) * 2005-12-30 2007-06-30 Centocor, Inc. A method for determining the phenotype of cells
US8283160B2 (en) 2007-09-11 2012-10-09 Frey Ii William H Methods, pharmaceutical compositions and articles of manufacture for administering therapeutic cells to the animal central nervous system
AU2008308531B2 (en) * 2007-10-05 2014-04-24 Ethicon, Incorporated Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
EP2379089B1 (de) * 2008-12-19 2019-04-17 DePuy Synthes Products, Inc. Regeneration und reparatur von nervengeweben nach einer verletzung
US10557116B2 (en) * 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
CN102498204B (zh) * 2009-03-26 2015-02-04 德普伊新特斯产品有限责任公司 人脐带组织细胞作为用于阿尔茨海默病的疗法
AU2012358810B2 (en) 2011-12-23 2018-03-15 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue-derived cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
JP6301263B2 (ja) 2018-03-28
KR20140133502A (ko) 2014-11-19
AU2012358810B2 (en) 2018-03-15
US20130190201A1 (en) 2013-07-25
ES2676556T3 (es) 2018-07-20
US9611513B2 (en) 2017-04-04
HK1213596A1 (zh) 2016-07-08
ZA201405407B (en) 2016-05-25
HK1203550A1 (en) 2015-10-30
PL2794854T3 (pl) 2018-12-31
AU2012358810A1 (en) 2014-08-07
CA2869681A1 (en) 2013-06-27
RU2636220C2 (ru) 2017-11-21
MX362198B (es) 2019-01-08
US20170335409A1 (en) 2017-11-23
US10724105B2 (en) 2020-07-28
WO2013096686A1 (en) 2013-06-27
BR112014015424A2 (pt) 2018-05-22
KR101981450B1 (ko) 2019-08-28
CN104837987A (zh) 2015-08-12
RU2014130221A (ru) 2016-02-20
JP2015504662A (ja) 2015-02-16
PH12014501445B1 (en) 2014-10-08
CN104837987B (zh) 2018-10-02
EP2794854A1 (de) 2014-10-29
MX2014007638A (es) 2015-05-15
SG11201403465PA (en) 2014-10-30
PH12014501445A1 (en) 2014-10-08

Similar Documents

Publication Publication Date Title
US10724105B2 (en) Detection of human umbilical cord tissue-derived cells
US10179900B2 (en) Conditioned media and methods of making a conditioned media
AU2019202191B2 (en) Nutrient enriched media for hUTC growth
EP1971681B1 (de) Produkte und methoden zu verhinderung schädlicher immunantworten bei histo-incompatiblen transplantationen
RU2748057C2 (ru) Композиция и способы криоконсервации клеток hutc
WO2008146991A1 (en) Process for the isolation of placenta-derived trophoblast stem cells

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20140722

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1203550

Country of ref document: HK

17Q First examination report despatched

Effective date: 20151116

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20171201

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: DEPUY SYNTHES PRODUCTS, INC.

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602012047706

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1010618

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180715

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2676556

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20180720

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180920

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180920

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180921

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1010618

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181020

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602012047706

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20190321

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181220

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20181231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180620

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180620

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20121220

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20210111

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20211028

Year of fee payment: 10

Ref country code: DE

Payment date: 20211102

Year of fee payment: 10

Ref country code: FR

Payment date: 20211115

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20211110

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20211103

Year of fee payment: 10

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20230329

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20211221

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602012047706

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20221220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221220

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230701

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221220